1
|
Al-Kadi A, Anter AF, Rofaeil RR, Sayed-Ahmed MM, Hafez SMNA, Ahmed ASF. Endothelin System Blockade Extenuates Sepsis-Induced Acute Heart and Kidney Injuries via Modulating ET-1/Klotho/p38-MAPK. Clin Exp Pharmacol Physiol 2025; 52:e70042. [PMID: 40228821 DOI: 10.1111/1440-1681.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Sepsis-induced organ failure is a major health problem, characterised by massive inflammatory and oxidative stress responses. Endothelin-1 (ET-1) is one of the peptides expressed during septicemia with proapoptotic, proinflammatory, and oxidant effects. ET-1 plays a role in heart and kidney injuries in sepsis. Accordingly, the current study was conducted to investigate, on a mechanistic basis, whether inhibition of ET-1 signalling either by blocking its receptors or inhibiting its formation attenuates sepsis-induced acute cardiorenal injuries. To analyse the role of ET-1 in sepsis, we used a cecal ligation and puncture (CLP) model of sepsis. The animals were divided into five groups: CLP non-treated group, CLP-treated groups with bosentan, ambrisentan, and phosphoramidon (30, 5, and 0.5 mg/kg, respectively), and sham-operated group. In addition to the same set of groups, survival analysis was assigned Survival rate, histopathological assessment, and cardiorenal functions were analysed. Oxidant and antioxidant activities, ET-1, IL-6, and lactate were measured. The expression of TNF-α, p38, Klotho, and caspase-3 was evaluated by immunohistochemistry. CLP caused acute cardiorenal damage, high mortality, upregulated levels of ET-1, IL-6, and lactate, as well as an imbalance in oxidant/antioxidant activities, elevated expression of TNF-α, p38, caspase-3 and reduced expression of klotho. Bosentan, ambrisentan, or phosphoramidon improved survival, reduced the levels of inflammatory and oxidative stress parameters, improved cardiorenal functions and structure, elevated the tissue contents of GSH and SOD, raised the expression of klotho protein, and reduced the cardiorenal expression of p38, TNF-α and caspase-3. Endothelin receptor antagonists (ERAs); bosentan and ambrisentan, or endothelin converting enzyme inhibitor (ECE-i) phosphoramidon, are promising agents against sepsis-induced organ damage. This was evident in their cardiorenal protective effects, up-regulation of klotho, suppression of inflammation, oxidation, apoptosis, and enhancement of the antioxidant status.
Collapse
Affiliation(s)
- Alaa Al-Kadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Aliaa F Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Remon Roshdy Rofaeil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, Egypt
- Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Mohamed M Sayed-Ahmed
- Pharmacology and Experimental Oncology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sara Mohamed Naguib Abdel Hafez
- Histology and Cell Biology Department, Minia University, Faculty of Medicine, Minia University Faculty of Medicine, Minia, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
2
|
Han YY, Yang JL, Meng HM, Wang P. Identifying Risk Factors for Myocardial Injury in Elderly Patients with Sepsis. Med Sci Monit 2025; 31:e947840. [PMID: 40382677 PMCID: PMC12101096 DOI: 10.12659/msm.947840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/04/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Myocardial injury is a common complication in elderly patients with sepsis and is associated with poor prognosis. This study aimed to identify clinical characteristics and independent risk factors for myocardial injury in elderly sepsis patients admitted to the Emergency Intensive Care Unit (EICU). MATERIAL AND METHODS A retrospective analysis was conducted on 160 elderly patients with sepsis admitted to the EICU, categorized into myocardial injury and non-myocardial injury groups. Demographic data, inflammatory markers, echocardiographic parameters, and blood urea nitrogen-to-albumin ratio (BAR) values were compared. Logistic regression identified independent risk factors, and ROC curve analysis assessed the predictive value of BAR. RESULTS Of 160 patients, 106 (63.1%) had myocardial injury, with an average age of 77.56±7.49 years. Myocardial injury was associated with lower ejection fraction (EF), and elevated procalcitonin, lactate, and BAR levels (P<0.05). Logistic regression identified septic shock (RR=2.612, P=0.003), elevated BAR (RR=2.272, P=0.035) and lactate levels (RR=1.145, P=0.010) as independent risk factors for myocardial injury. In contrast, increased EF (RR=0.932, P=0.007) was identified as protective against myocardial injury, with lower EF associated with a higher risk. ROC analysis showed that BAR had moderate predictive value (AUC=0.653, P<0.01), with sensitivity of 76.4% and specificity of 53.2% at an optimal cutoff of 0.33. CONCLUSIONS Septic shock, reduced EF, and elevated BAR and lactate levels are independent risk factors for myocardial injury in elderly patients with sepsis. BAR serves as an early marker for myocardial injury, aiding in risk assessment and management in the EICU.
Collapse
|
3
|
Xue J, Zhou N, Li Q, Wang R, Li Y, Zhu H, Lv C. Exploring the pathogenesis of sepsis-induced cardiomyopathy: Multilayered mechanisms and clinical responses. Sci Prog 2025; 108:368504251329190. [PMID: 40112325 PMCID: PMC11926820 DOI: 10.1177/00368504251329190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Sepsis-induced cardiomyopathy (SIC), as a common complication in the intensive care unit, not only increases the complexity of patient care but also greatly enhances the risk of death. Currently, clinical management of SIC remains challenging, mainly due to the complexity of its pathogenesis and the lack of targeted therapies. Although the specific etiology of SIC is not yet fully understood, existing studies have revealed several vital pathological processes that are intertwined and contribute to the progression of the disease. This narrative review summarizes the existing pathogenesis of SIC, which involves multiple aspects including the inflammatory response, mitochondrial dysfunction, cell death mechanisms, immune regulation, and calcium homeostasis imbalance. Given the multifactorial pathogenesis of SIC, future studies need to explore the interactions between these mechanisms and how to intervene to develop more precise and effective therapeutic strategies to reduce mortality and improve prognosis in patients with SIC.
Collapse
Affiliation(s)
- Jinfang Xue
- Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
| | - Ning Zhou
- Emergency Department, Central People's Hospital of Zhanjiang, Chikan District, Zhanjiang, Guangdong, China
| | - Quan Li
- Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
| | - Ruijie Wang
- Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
| | - Yan Li
- Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huadong Zhu
- Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chuanzhu Lv
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| |
Collapse
|
4
|
Meng X, Yan X, Xue P, Xi Z. Xuebijing Exerts Protective Effects on Myocardial Cells by Upregulating TRIM16 and Inhibiting Oxidative Stress and Apoptosis. Curr Comput Aided Drug Des 2025; 21:503-516. [PMID: 39623713 DOI: 10.2174/0115734099318323241122184120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/09/2024] [Accepted: 10/19/2024] [Indexed: 05/28/2025]
Abstract
OBJECTIVE This study utilized transcriptomic sequencing combined with cellular and animal models to explore the potential mechanisms of Xuebijing in treating sepsis-induced myocardial dysfunction, also known as sepsis-induced myocardial injury. METHODS We investigated potential targets and regulatory mechanisms of XBJ injection using network pharmacology and RNA sequencing. The effects of XBJ on oxidative stress and apoptosis levels in human cardiac myocytes (AC16) and C57BL/6 mice exposed to lipopolysaccharide (LPS) were evaluated by Enzyme-Linked Immunosorbent Assay (ELISA), fluorescent probe, Fluorescent Quantitative Polymerase Chain Reaction (qPCR), Western Blot, Transmission Electron Microscopy, oxidative stress-related indicators detection kit, flow cytometry, and Immunohistochemistry (IHC). RESULTS First, it was verified that XBJ can reduce the deformation of AC16 cardiomyocytes induced by LPS and the production and secretion of ROS (P <0.01). The transcriptome sequencing results showed that the TRIM16 gene was significantly increased after XBJ treatment, and the data of KEGG and GO analyses demonstrated that XBJ could inhibit the pathway expression of oxidative stress damage in AC16 cells, and PCR verified that XBJ could indeed increase the expression level of TRIM16 gene in AC16 cells (P <0.01). Basic animal and cell experiments showed that LPS could inhibit the expression of TRIM16 and NRF2 in cardiomyocytes (P <0.05) and promote the expression of Keap1 (P <0.01), while XBJ could significantly upregulate the expression levels of TRIM16 and NRF2 (P <0.01) and inhibit the expression of Keap1 (P <0.01), thereby affecting the expression levels of downstream proinflammatory cytokines and alleviating LPS-induced oxidative stress damage. In addition, XBJ also inhibited the expression of the pro-apoptotic proteins Bax and c-caspase3 (P <0.01), promoted the expression of the anti-apoptotic protein Bcl2 (P <0.01), and reduced LPS-induced apoptosis by upregulating TRIM16. CONCLUSION Our comprehensive data demonstrated that TRIM16 is a key gene in the therapeutic action of Xuebijing in sepsis-induced myocardial dysfunction, protecting myocardial cells from injury through antioxidative stress and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Xiaoyan Meng
- Department of Emergency Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210002, PR China
- Department of Intensive Care Unit, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 210000, PR China
| | - Xinming Yan
- Department of Emergency Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210002, PR China
- Department of Emergency Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 210000, PR China
| | - Peng Xue
- Department of Emergency Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210002, PR China
- Department of Intensive Care Unit, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 210000, PR China
| | - Zhaoqing Xi
- Department of Emergency Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210002, PR China
| |
Collapse
|
5
|
Miao S, Liu H, Yang Q, Zhang Y, Chen T, Chen S, Mao X, Zhang Q. Cathelicidin peptide LL-37: A multifunctional peptide involved in heart disease. Pharmacol Res 2024; 210:107529. [PMID: 39615616 DOI: 10.1016/j.phrs.2024.107529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/30/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Heart disease is a common human disease with high morbidity and mortality. Timely and effective prevention and treatment is an urgent clinical problem. The pathogenesis of heart disease is complex and diverse, involving hypertension, diabetes, atherosclerosis, drug toxicity, thrombosis, infection and other aspects. LL-37, an endogenous peptide, is well known for its antimicrobial properties. In recent years, LL-37 has been found to have a variety of biological functions, including its role in the regulation of atherosclerosis, thrombosis, inflammatory responses, and cardiac hypertrophy. Engineered LL-37-related peptides were developed and proved to regulate the development of disease, which revealed its potential clinical application. A comprehensive review and summary of LL-37 is presented to clarify its role in heart disease and to provide a reference and direction for future research.
Collapse
Affiliation(s)
- Shuo Miao
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China; School of Basic Medicine, Qingdao University, Qingdao, China
| | - Houde Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qingyu Yang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaping Zhang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Chen
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Ruipule Medical Technology Co., Ltd, China
| | - Shuai Chen
- School of Basic Medicine, Guizhou University of Traditional Chinese, China
| | - Xin Mao
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Qingsong Zhang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
6
|
Li H, Li X, Xu G, Zhan F. Minocycline alleviates lipopolysaccharide-induced cardiotoxicity by suppressing the NLRP3/Caspase-1 signaling pathway. Sci Rep 2024; 14:21180. [PMID: 39261543 PMCID: PMC11390881 DOI: 10.1038/s41598-024-72133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
Minocycline (Min), as an antibiotic, possesses various beneficial properties such as anti-inflammatory, antioxidant, and anti-apoptotic effects. Despite these known qualities, the precise cardioprotective effect and mechanism of Min in protecting against sepsis-induced cardiotoxicity (SIC) remain unspecified. To address this, our study sought to assess the protective effects of Min on the heart. Lipopolysaccharide (LPS) was utilized to establish a cardiotoxicity model both in vivo and in vitro. Min was pretreated in the models. In the in vivo setting, evaluation of heart tissue histopathological injury was performed using hematoxylin and eosin (H&E) staining and TUNEL. Immunohistochemistry (IHC) was employed to evaluate the expression levels of NLRP3 and Caspase-1 in the heart tissue of mice. During in vitro experiments, the viability of H9c2 cells was gauged utilizing the CCK8 assay kit. Intracellular ROS levels in H9c2 cells were quantified using a ROS assay kit. Both in vitro and in vivo settings were subjected to measurement of oxidative stress indexes, encompassing glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) levels. Additionglly, myocardial injury markers like lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB) activity were quantified using appropriate assay kits. Western blotting (WB) analysis was conducted to detect the expression levels of NOD-like receptor protein-3 (NLRP3), caspase-1, IL-18, and IL-1β, alongside apoptosis-related proteins such as Bcl-2 and Bax, and antioxidant proteins including superoxide dismutase-1 (SOD-1) and antioxidant proteins including superoxide dismutase-1 (SOD-2), both in H9c2 cells and mouse heart tissues. In vivo, Min was effective in reducing LPS-induced inflammation in cardiac tissue, preventing cell damage and apoptosis in cardiomyocytes. The levels of LDH and CK-MB were significantly reduced with Min treatment. In vitro studies showed that Min improved the viability of H9C2 cells, reduced apoptosis, and decreased ROS levels in these cells. Further analysis indicated that Min decreased the protein levels of NLRP3, Caspase-1, IL-18, and IL-1β, while increasing the levels of SOD-1 and SOD-2 both in vivo and in vitro. Min alleviates LPS-induced SIC by suppressing the NLRP3/Caspase-1 signalling pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Huijuan Li
- Department of Anesthesiology, Wuhan Third Hospital, Wuhan, 430074, China
| | - Xiaozhong Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China.
| |
Collapse
|
7
|
Lian H, Li S, Zhang Q, Wang X, Zhang H. U-shaped prognostic value of left ventricular-arterial coupling in septic patients: a prospective study. Eur J Med Res 2024; 29:435. [PMID: 39210468 PMCID: PMC11360502 DOI: 10.1186/s40001-024-02037-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Ventricular-arterial coupling (VAC) has garnered increasing interest in critical care. The prognostic significance of left ventricular-arterial coupling (LVAC) in this context remains a topic of debate. OBJECTIVE This study aimed to explore the association between LVAC and patient outcomes in sepsis. METHODS Patients with sepsis or septic shock admitted to the intensive care unit (ICU) were included. LVAC was evaluated using the arterial elastance (Ea)/left ventricular end-systolic elastance (Ees) ratio. Prognostic indicators, including 30-day mortality, length of ICU stay, mechanical ventilation (MV), changes in delta lactate levels, and oxygen index were also collected. RESULTS A total of 388 patients were enrolled in this study. A U-shaped relationship was observed between LVAC and 30-day mortality, with an optimal LVAC value of 1.19 identified. For LVAC values above 1.19, the odds ratio (OR) for 30-day mortality was 1.07 (95% confidence interval [CI] 1.01-1.14). Below this threshold, OR was 0.85 (95% CI 0.73, 0.99). Similarly, in the curve for ICU-free days, a β value of - 8.64 (95% CI - 16.53, - 0.76) was noted for LVAC values over 1.26. For ventilator-free time, the kink point was 1.24, with significant β values on both sides of this threshold [- 226.49 (95% CI - 411.59, - 41.38) and 147.67 (95% CI 12.40, 282.93), respectively]. CONCLUSIONS This study established U-shaped associations between LVAC and various clinical outcomes in septic patients. Optimizing LVAC could potentially enhance patient prognosis. Given the slight variations in optimal LVAC values across different patient populations, individualized LVAC titration may be beneficial in improving clinical outcomes.
Collapse
Affiliation(s)
- Hui Lian
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Suwei Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qing Zhang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongmin Zhang
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Zhang Y, Li S, Fan X, Wu Y. Pretreatment with Indole-3-Propionic Acid Attenuates Lipopolysaccharide-Induced Cardiac Dysfunction and Inflammation Through the AhR/NF-κB/NLRP3 Pathway. J Inflamm Res 2024; 17:5293-5309. [PMID: 39157586 PMCID: PMC11330251 DOI: 10.2147/jir.s466777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Background Patients with sepsis frequently develop septic cardiomyopathy, which is known to be closely related to excessive inflammatory responses. Indole-3-propionic acid (IPA) is a tryptophan metabolite with anti-inflammatory properties that have been demonstrated in various studies. In this study, we investigated the underlying mechanisms and therapeutic role of IPA in septic cardiomyopathy. Methods To investigate the role of IPA in septic cardiomyopathy, we constructed a lipopolysaccharide (LPS)-induced rat model of septic cardiomyopathy, and treated rats with IPA. Inflammatory factors and the NF-κB/NLRP3 pathway were evaluated in myocardial tissues and cells after IPA treatment using RT-qPCR, ELISA, Western blotting, and immunohistochemistry. To further elucidate the role of the aryl hydrocarbon receptor (AhR), we detected changes in inflammatory mediators and the NF-κB/NLRP3 pathway in in vivo and in vitro models of septic cardiomyopathy, which were treated with the AhR antagonist CH-223191 and/or AhR agonist FICZ. Results IPA supplementation improved cardiac dysfunction in rats with septic cardiomyopathy. IPA reduced inflammatory cytokine release and inhibited NF-κB/NLRP3 signaling pathway in myocardial tissue and in H9c2 cells. CH-223191 impaired the anti-inflammatory effect of IPA in LPS-treated cells, whereas FICZ exerted the same effect as IPA. IPA also exhibited anti-inflammatory activity by binding to the AhR. Our results indicated that IPA attenuated septic cardiomyopathy in rats via AhR/NF-κB/NLRP3 signaling. Conclusion Our study revealed that IPA improved left heart dysfunction and myocardial inflammation caused by sepsis via AhR/NF-κB/NLRP3 signaling, suggesting that IPA is a potential therapy for septic cardiomyopathy.
Collapse
Affiliation(s)
- Yiqiong Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Shanshan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xiaojuan Fan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
9
|
Casper E, El Wakeel L, Sabri N, Khorshid R, Fahmy SF. Melatonin: A potential protective multifaceted force for sepsis-induced cardiomyopathy. Life Sci 2024; 346:122611. [PMID: 38580195 DOI: 10.1016/j.lfs.2024.122611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Sepsis is a life-threatening condition manifested by organ dysfunction caused by a dysregulated host response to infection. Lung, brain, liver, kidney, and heart are among the affected organs. Sepsis-induced cardiomyopathy is a common cause of death among septic patients. Sepsis-induced cardiomyopathy is characterized by an acute and reversible significant decline in biventricular both systolic and diastolic function. This is accompanied by left ventricular dilatation. The pathogenesis underlying sepsis-induced cardiomyopathy is multifactorial. Hence, targeting an individual pathway may not be effective in halting the extensive dysregulated immune response. Despite major advances in sepsis management strategies, no effective pharmacological strategies have been shown to treat or even reverse sepsis-induced cardiomyopathy. Melatonin, namely, N-acetyl-5-methoxytryptamine, is synthesized in the pineal gland of mammals and can also be produced in many cells and tissues. Melatonin has cardioprotective, neuroprotective, and anti-tumor activity. Several literature reviews have explored the role of melatonin in preventing sepsis-induced organ failure. Melatonin was found to act on different pathways that are involved in the pathogenesis of sepsis-induced cardiomyopathy. Through its antimicrobial, anti-inflammatory, and antioxidant activity, it offers a potential role in sepsis-induced cardiomyopathy. Its antioxidant activity is through free radical scavenging against reactive oxygen and nitrogen species and modulating the expression and activity of antioxidant enzymes. Melatonin anti-inflammatory activities control the overactive immune system and mitigate cytokine storm. Also, it mitigates mitochondrial dysfunction, a major mechanism involved in sepsis-induced cardiomyopathy, and thus controls apoptosis. Therefore, this review discusses melatonin as a promising drug for the management of sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Eman Casper
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Lamia El Wakeel
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Nagwa Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Ramy Khorshid
- Department of Cardiovascular and Thoracic Surgery, Ain Shams University Hospital, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Sarah F Fahmy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
10
|
Lee KJ, Kim YK, Jeon K, Ko RE, Suh GY, Oh DK, Lim SY, Lee YJ, Lee SY, Park MH, Lim CM, Park S. Shock indices are associated with in-hospital mortality among patients with septic shock and normal left ventricular ejection fraction. PLoS One 2024; 19:e0298617. [PMID: 38470900 DOI: 10.1371/journal.pone.0298617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/27/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The understanding of shock indices in patients with septic shock is limited, and their values may vary depending on cardiac function. METHODS This prospective cohort study was conducted across 20 university-affiliated hospitals (21 intensive care units [ICUs]). Adult patients (≥19 years) with septic shock admitted to the ICUs during a 29-month period were included. The shock index (SI), diastolic shock index (DSI), modified shock index (MSI), and age shock index (Age-SI) were calculated at sepsis recognition (time zero) and ICU admission. Left ventricular (LV) function was categorized as either normal LV ejection fraction (LVEF ≥ 50%) or decreased LVEF (<50%). RESULTS Among the 1,194 patients with septic shock, 392 (32.8%) who underwent echocardiography within 24 h of time zero were included in the final analysis (normal LVEF: n = 246; decreased LVEF: n = 146). In patients with normal LVEF, only survivors demonstrated significant improvement in SI, DSI, MSI, and Age-SI values from time zero to ICU admission; however, no notable improvements were found in all patients with decreased LVEF. The completion of vasopressor or fluid bundle components was significantly associated with improved indices in patients with normal LVEF, but not in those with decreased LVEF. In multivariable analysis, each of the four indices at ICU admission was significantly associated with in-hospital mortality (P < 0.05) among patients with normal LVEF; however, discrimination power was better in the indices for patients with lower lactate levels (≤ 4.0 mmol/L), compared to those with higher lactate levels. CONCLUSIONS The SI, DSI, MSI, and Age-SI at ICU admission were significantly associated with in-hospital mortality in patients with septic shock and normal LVEF, which was not found in those with decreased LVEF. Our study emphasizes the importance of interpreting shock indices in the context of LV function in septic shock.
Collapse
Affiliation(s)
- Kyu Jin Lee
- Department of Pulmonary, Allergy and Critical Care Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Yong Kyun Kim
- Department of Infection, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Kyeongman Jeon
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ryoung-Eun Ko
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Gee Young Suh
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Kyu Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Yoon Lim
- Department of Pulmonary and Critical Care Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yeon Joo Lee
- Department of Pulmonary and Critical Care Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Su Yeon Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi-Hyeon Park
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chae-Man Lim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sunghoon Park
- Department of Pulmonary, Allergy and Critical Care Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| |
Collapse
|
11
|
Cheng L, Liang J, Xie F, Han Z, Luo W, Chen H, He J. Identification and validation of a novel glycolysis-related ceRNA network for sepsis-induced cardiomyopathy. Front Med (Lausanne) 2024; 11:1343281. [PMID: 38439898 PMCID: PMC10910075 DOI: 10.3389/fmed.2024.1343281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Purpose Sepsis-induced cardiomyopathy (SIC) is a major life-threatening condition in critically infected patients. Early diagnosis and intervention are important to improve patient prognosis. Recognizing the pivotal involvement of the glycolytic pathway in SIC, this study aims to establish a glycolysis-related ceRNA network and explore novel diagnostic avenues. Materials and methods SIC-related datasets were carefully filtered from the GEO database. CytoHubba was used to identify differentially expressed genes (DEGs) associated with glycolysis. A predictive method was then used to construct an lncRNA-miRNA-mRNA network. Dual-luciferase reporter assays validated gene interactions, and the specificity of this ceRNA network was confirmed in peripheral blood mononuclear cells (PBMCs) from SIC patients. Logistic analysis was used to examine the correlation between the ceRNA network and SIC. Diagnostic potential was assessed using receiver operating characteristic (ROC) curves, and correlation analysis investigated any associations between gene expression and clinical indicators. Results IER3 was identified as glycolysis-related DEG in SIC, and a ceRNA network (SNHG17/miR-214-3p/IER3) was established by prediction. Dual luciferase reporter gene assay confirmed the presence of mutual binding between IER3, miR-214-3p and SNHG17. RT-qPCR verified the specific expression of this ceRNA network in SIC patients. Multivariate logistic analysis established the correlation between the ceRNA network and SIC. ROC analysis demonstrated its high diagnostic specificity (AUC > 0.8). Correlation analysis revealed a negative association between IER3 expression and oxygenation index in SIC patients (p < 0.05). Furthermore, miR-214-3p expression showed a negative correlation with NT-proBNP (p < 0.05). Conclusion In this study, we identified and validated a ceRNA network associated with glycolysis in SIC: SNHG17/miR-214-3p/IER3. This ceRNA network may play a critical role in the onset and development of SIC. This finding is important to further our understanding of the pathophysiological mechanisms underlying SIC and to explore potential diagnostic and therapeutic targets for SIC.
Collapse
Affiliation(s)
- Lulu Cheng
- Postgraduate Cultivation Base of Guangzhou University of Chinese Medicine, Panyu Central Hospital, Guangzhou, China
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jiabin Liang
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Zeping Han
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Hanwei Chen
- Radiology Department of Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou, China
| | - Jinhua He
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| |
Collapse
|
12
|
Geng H, Zhang H, Cheng L, Dong S. Sivelestat ameliorates sepsis-induced myocardial dysfunction by activating the PI3K/AKT/mTOR signaling pathway. Int Immunopharmacol 2024; 128:111466. [PMID: 38176345 DOI: 10.1016/j.intimp.2023.111466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 12/07/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
The cardioprotective role of sivelestat, a neutrophil elastase inhibitor, has already been demonstrated, but the underlying molecular mechanism remains unclear. This study aimed to explore the mechanism underlying the role of sivelestat in sepsis-induced myocardial dysfunction (SIMD). We found that sivelestat treatment remarkably improved the viability and suppressed the apoptosis of lipopolysaccharide (LPS)-stimulated H9c2 cells. In vivo, sivelestat treatment was associated with an improved survival rate; reduced serum cTnT, TNF-α, IL-1β levels and myocardial TNF-α and IL-1β levels; ameliorated cardiac function and structure; and reduced cardiomyocyte apoptosis. Moreover, sivelestat treatment substantially increased Bcl-2 expression and suppressed caspase-3 and Bax expression in LPS-induced H9c2 cells and in the heart tissues of septic rats. Furthermore, the phosphatidylinositol 3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) signaling pathway was activated both in vitro and in vivo. The protective effect of sivelestat against SIMD was reversed by the PI3K inhibitor LY294002. In summary, sivelestat can protect against SIMD by activating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hongyu Geng
- Department of Intensive Care Unit, Baoding First Central Hospital, Baoding, China
| | - Hongbo Zhang
- Department of General Surgery, Baoding First Central Hospital, Baoding, China
| | - Lianfang Cheng
- Department of Intensive Care Unit, Baoding First Central Hospital, Baoding, China
| | - Shimin Dong
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
13
|
Li X, Zhang Z, Zhang X, Yin Y, Yuan X, You X, Wu J. Echinacoside Prevents Sepsis-Induced Myocardial Damage via Targeting SOD2. J Med Food 2024; 27:123-133. [PMID: 38100058 DOI: 10.1089/jmf.2023.k.0222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024] Open
Abstract
Echinacoside (ECH) is a prominent naturally occurring bioactive compound with effects of alleviating myocardial damage. We aimed to explore the beneficial effects of ECH against sepsis-induced myocardial damage and elucidate the potential mechanism. Echocardiography and Masson staining demonstrated that ECH alleviates cardiac function and fibrosis in the cecal ligation and puncture (CLP) model. Transcriptome profiling and network pharmacology analysis showed that there are 51 overlapping targets between sepsis-induced myocardial damage and ECH. Subsequently, chemical carcinogenesis-reactive oxygen species (ROS) were enriched in multiple targets. Wherein, SOD2 may be the potential target of ECH on sepsis-induced myocardial damage. Polymerase chain reaction results showed that ECH administration could markedly increase the expression of SOD2 and reduce the release of ROS. Combined with injecting the inhibitor of SOD2, the beneficial effect of ECH on mortality, cardiac function, and fibrosis was eliminated, and release of ROS was increased after inhibiting SOD2. ECH significantly alleviated myocardial damage in septic mice, and the therapeutic mechanism of ECH is achieved by upregulating SOD2 which decreased the release of ROS.
Collapse
Affiliation(s)
- Xin Li
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology (USST), Shanghai, China
| | - Zuojing Zhang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiaoxuan Zhang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yibo Yin
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinru Yuan
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xingji You
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology (USST), Shanghai, China
| |
Collapse
|
14
|
Liu D, Wang T, Wang Q, Dong P, Liu X, Li Q, Shi Y, Li J, Zhou J, Zhang Q. Identification of key genes in sepsis-induced cardiomyopathy based on integrated bioinformatical analysis and experiments in vitro and in vivo. PeerJ 2023; 11:e16222. [PMID: 38025678 PMCID: PMC10668858 DOI: 10.7717/peerj.16222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Sepsis is a life-threatening disease that damages multiple organs and induced by the host's dysregulated response to infection with high morbidity and mortality. Heart remains one of the most vulnerable targets of sepsis-induced organ damage, and sepsis-induced cardiomyopathy (SIC) is an important factor that exacerbates the death of patients. However, the underlying genetic mechanism of SIC disease needs further research. Methods The transcriptomic dataset, GSE171564, was downloaded from NCBI for further analysis. Gene expression matrices for the sample group were obtained by quartile standardization and log2 logarithm conversion prior to analysis. The time series, protein-protein interaction (PPI) network, and functional enrichment analysis via Gene Ontology and KEGG Pathway Databases were used to identify key gene clusters and their potential interactions. Predicted miRNA-mRNA relationships from multiple databases facilitated the construction of a TF-miRNA-mRNA regulatory network. In vivo experiments, along with qPCR and western blot assays, provided experimental validation. Results The transcriptome data analysis between SIC and healthy samples revealed 221 down-regulated, and 342 up-regulated expressed genes across two distinct clusters. Among these, Tpt1, Mmp9 and Fth1 were of particular significance. Functional analysis revealed their role in several biological processes and pathways, subsequently, in vivo experiments confirmed their overexpression in SIC samples. Notably, we found TPT1 play a pivotal role in the progression of SIC, and silencing TPT1 showed a protective effect against LPS-induced SIC. Conclusion In our study, we demonstrated that Tpt1, Mmp9 and Fth1 have great potential to be biomarker of SIC. These findings will facilitated to understand the occurrence and development mechanism of SIC.
Collapse
Affiliation(s)
- Dehua Liu
- Weifang Medical University, Weifang, China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qingguo Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Peikang Dong
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaohong Liu
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qiang Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Youkui Shi
- Department of Emergency Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jin Zhou
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Quan Zhang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
15
|
Taha AM, Mahmoud AM, Ghonaim MM, Kamran A, AlSamhori JF, AlBarakat MM, Shrestha AB, Jaiswal V, Reiter RJ. Melatonin as a potential treatment for septic cardiomyopathy. Biomed Pharmacother 2023; 166:115305. [PMID: 37619482 DOI: 10.1016/j.biopha.2023.115305] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Septic cardiomyopathy (SCM) is a common complication of sepsis contributing to high mortality rates. Its pathophysiology involves complex factors, including inflammatory cytokines, mitochondrial dysfunction, oxidative stress, and immune dysregulation. Despite extensive research, no effective pharmacological agent has been established for sepsis-induced cardiomyopathy. Melatonin, a hormone with diverse functions in the body, has emerged as a potential agent for SCM through its anti-oxidant, anti-inflammatory, anti-apoptotic, and cardioprotective roles. Through various molecular levels of its mechanism of action, it counterattacks the adverse event of sepsis. Experimental studies have mentioned that melatonin protects against many cardiovascular diseases and exerts preventive effects on SCM. Moreover, melatonin has been investigated in combination with other drugs such as antibiotics, resveratrol, and anti-oxidants showing synergistic effects in reducing inflammation, anti-oxidant, and improving cardiac function. While preclinical studies have demonstrated positive results, clinical trials are required to establish the optimal dosage, route of administration, and treatment duration for melatonin in SCM. Its safety profile, low toxicity, and natural occurrence in the human body provide a favorable basis for its clinical use. This review aims to provide an overview of the current evidence of the use of melatonin in sepsis-induced cardiomyopathy (SICM). Melatonin appears to be promising as a possible treatment for sepsis-induced cardiomyopathy and demands further investigation.
Collapse
Affiliation(s)
- Amira Mohamed Taha
- Faculty of Medicine, Fayoum University, Fayoum, Egypt; Medical Research Group of Egypt (MRGE), Negida Academy, Arlington, MA, USA
| | | | | | - Ateeba Kamran
- Bachelor of Medicine, Bachelor of Surgery, Karachi Medical and Dental College, Karachi, Pakistan
| | | | - Majd M AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Abhigan Babu Shrestha
- Department of Internal Medicine, M Abdur Rahim Medical College, Dinajpur, Bangladesh.
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
16
|
Bansal M, Mehta A, Machanahalli Balakrishna A, Kalyan Sundaram A, Kanwar A, Singh M, Vallabhajosyula S. RIGHT VENTRICULAR DYSFUNCTION IN SEPSIS: AN UPDATED NARRATIVE REVIEW. Shock 2023; 59:829-837. [PMID: 36943772 DOI: 10.1097/shk.0000000000002120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
ABSTRACT Sepsis is a multisystem disease process, which constitutes a significant public health challenge and is associated with high morbidity and mortality. Among other systems, sepsis is known to affect the cardiovascular system, which may manifest as myocardial injury, arrhythmias, refractory shock, and/or septic cardiomyopathy. Septic cardiomyopathy is defined as the reversible systolic and/or diastolic dysfunction of one or both ventricles. Left ventricle dysfunction has been extensively studied in the past, and its prognostic role in patients with sepsis is well documented. However, there is relatively scarce literature on right ventricle (RV) dysfunction and its role. Given the importance of timely detection of septic cardiomyopathy and its bearing on prognosis of patients, the role of RV dysfunction has come into renewed focus. Hence, through this review, we sought to describe the pathophysiology of RV dysfunction in sepsis and what have we learnt so far about its multifactorial nature. We also elucidate the roles of different biomarkers for its detection and prognosis, along with appropriate management of such patient population.
Collapse
Affiliation(s)
- Mridul Bansal
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Aryan Mehta
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | | | - Arvind Kalyan Sundaram
- Section of Cardiovascular Medicine, Department of Medicine, UMass Chan-Baystate Medical Center, Springfield, Massachusetts
| | | | - Mandeep Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
17
|
Lima MR, Silva D. Septic cardiomyopathy: A narrative review. Rev Port Cardiol 2023; 42:471-481. [PMID: 36893835 DOI: 10.1016/j.repc.2021.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/03/2021] [Accepted: 05/01/2021] [Indexed: 03/09/2023] Open
Abstract
Sepsis is a systemic inflammatory response syndrome of suspected or documented infectious origin, whose outcome is multiorgan failure. Sepsis-induced myocardial dysfunction (SIMD), present in more than 50% of septic patients, is characterized by (i) left ventricular (LV) dilatation with normal or low filling pressure, (ii) right and/or LV (systolic and/or diastolic) dysfunction and (iii) reversibility. Since the first definition proposed by Parker et al. in 1984, attempts have been made to define SIMD. Many parameters are used to assess cardiac function in septic patients, sometimes making it more difficult to measure due to the intrinsic hemodynamical changes in this condition. Nevertheless, with advanced echocardiographic techniques, such as speckle tracking analysis, it is possible to diagnose and assess systolic and diastolic dysfunction, even in the earliest stages of sepsis. Cardiac magnetic resonance imaging brings new insights into the reversibility of this condition. Many uncertainties still remain regarding the mechanisms, characteristics, treatment and even prognosis of this condition. There are also inconsistent conclusions from studies, therefore this review attempts to summarize our current knowledge of SIMD.
Collapse
Affiliation(s)
- Maria Rita Lima
- Internal Medicine Department, Egas Moniz Hospital, Lisbon Ocidental Hospital Center, Lisbon, Portugal.
| | - Doroteia Silva
- Intensive Care Department, Santa Maria University Hospital, Lisbon North Hospital Center, Lisbon, Portugal; CCUL, Lisbon Academic Medical Center, Faculty of Medicine of Lisbon, Lisbon, Portugal
| |
Collapse
|
18
|
Du Y, Zhong Y, Ding R, Wang X, Xia F, Zhang Q, Peng Q. New insights of necroptosis and immune infiltration in sepsis-induced myocardial dysfunction from bioinformatics analysis through RNA-seq in mice. Front Cell Infect Microbiol 2022; 12:1068324. [PMID: 36619743 PMCID: PMC9811394 DOI: 10.3389/fcimb.2022.1068324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by dysregulated host immune response to infection. Sepsis-induced myocardial dysfunction (SIMD) is a common complication in patients with severe sepsis and is associated with increased mortality. The molecular mechanisms underlying SIMD are complex and not well characterized. Excessive inflammation due to impaired regulation of immune response is one of the major causes of SIMD. Necroptosis is a novel type of cell death that is closely related to tissue injury and inflammation. However, the role of necroptosis in SIMD is not known. Therefore, in this study, we performed an in-depth bioinformatics analysis to investigate the relationship between necroptosis and SIMD using a mouse model generated by intraperitoneal injection of lipopolysaccharide (LPS) and the underlying mechanisms. Myocardial function was assessed by echocardiography. Histopathological changes in SIMD were analyzed by hematoxylin and eosin (H&E) staining. Gene expression profiles of the heart tissues from the SIMD and control mice were analyzed by bioinformatics analysis. Transcriptome sequencing demonstrated significant differences in the expression levels of 3654 genes in the heart tissues of SIMD mice including 1810 up-regulated and 1844 down-regulated genes. The necroptosis pathway genes were significantly enriched in the heart tissues from the SIMD group mice. We identified 35 necroptosis-related differentially expressed genes (NRDEGs) including MLKL and RIPK3. Cardiomyocyte necroptosis was confirmed by qRT-PCR and western blot analysis. The expression levels of most NRDEGs showed positive correlation with the infiltration levels of mast cells, macrophages, and neutrophils, and negative correlation with the infiltration levels of B cells and plasma cells in the heart tissues of the SIMD group mice. In conclusion, this study demonstrated that necroptosis was associated with changes in the infiltration levels of several immune cell types in the heart tissues of the SIMD model mice. This suggested that necroptosis influenced SIMD development by modulating the immune microenvironment. This suggested that NRDEGs are potential diagnostic biomarkers and therapeutic targets for patients with SIMD.
Collapse
Affiliation(s)
- Yan Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ruilin Ding
- Institute of Drug Clinical Trial/GCP Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaojie Wang
- Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fenfen Xia
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qian Zhang
- Department of Infectious Diseases, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China,*Correspondence: Qian Zhang, ; Qing Peng,
| | - Qing Peng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China,*Correspondence: Qian Zhang, ; Qing Peng,
| |
Collapse
|
19
|
Saito Y, Otaki Y, Watanabe T, Tachibana S, Sato J, Kobayashi Y, Aono T, Goto J, Wanezaki M, Kutsuzawa D, Kato S, Tamura H, Nishiyama S, Arimoto T, Takahashi H, Watanabe M. Cardiac-specific ITCH overexpression ameliorates septic cardiomyopathy via inhibition of the NF-κB signaling pathway. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 2:100018. [PMID: 39802494 PMCID: PMC11708253 DOI: 10.1016/j.jmccpl.2022.100018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 01/16/2025]
Abstract
Background Septic cardiomyopathy is a common complication of septic shock and organ dysfunction. ITCH is a HECT (homologous to the E6-AP carboxyl-terminus)-type ubiquitin E3 ligase that plays a critical role in inflammatory suppression. Herein, we focused on the interaction between ITCH and key regulators of nuclear factor-κB (NF-κB), such as tumor necrosis factor receptor-associated factor 6 (TRAF6) and transforming growth factor-β activated kinase 1 (TAK1), and examined the impact of ITCH on the development of septic cardiomyopathy. Methods and results In H9C2 cardiomyocytes, ITCH protein expression decreased in response to lipopolysaccharide (LPS) and tumor necrosis factor alpha (TNFα). The protein interactions of ITCH with TRAF6 and TAK1 were confirmed by immunoprecipitation in vitro and in vivo. Based on overexpression and knockdown studies of ITCH in H9C2 cardiomyocytes, ITCH regulates the phosphorylation of NF-κB and subsequent interleukin 6 (IL-6) expression in response to LPS and TNFα stimulation. LPS was intraperitoneally injected into transgenic mice with cardiac-specific overexpression of ITCH (ITCH-Tg) and wild-type (WT) mice. Compared with WT mice, phosphorylation of NF-κB and subsequent IL-6 expression were inhibited in ITCH-Tg mice. Cardiac systolic dysfunction after LPS administration was ameliorated in ITCH-Tg mice, and the survival rate was higher in ITCH-Tg mice than in WT mice. Conclusion ITCH interacts with TRAF6 and TAK1 in cardiomyocytes and improves cardiac function and survival rates in septic cardiomyopathy by suppressing the NF-κB pathway.
Collapse
Affiliation(s)
- Yuji Saito
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Yoichiro Otaki
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Shingo Tachibana
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Junya Sato
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Yuta Kobayashi
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Tomonori Aono
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Jun Goto
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Masahiro Wanezaki
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Daisuke Kutsuzawa
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Shigehiko Kato
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Harutoshi Tamura
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Satoshi Nishiyama
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Takanori Arimoto
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Hiroki Takahashi
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| |
Collapse
|
20
|
Zhu H, Zhang L, Jia H, Xu L, Cao Y, Zhai M, Li K, Xia L, Jiang L, Li X, Zhou Y, Liu J, Yu S, Duan W. Tetrahydrocurcumin improves lipopolysaccharide-induced myocardial dysfunction by inhibiting oxidative stress and inflammation via JNK/ERK signaling pathway regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154283. [PMID: 35779282 DOI: 10.1016/j.phymed.2022.154283] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/06/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Acute myocardial dysfunction in patients with sepsis is attributed to oxidative stress, inflammation, and cardiomyocyte loss; however, specific drugs for its prevention are still lacking. Tetrahydrocurcumin (THC) has been proven to contribute to the prevention of various cardiovascular diseases by decreasing oxidative stress and inflammation. This study was performed to investigate the functions and mechanism of action of THC in septic cardiomyopathy. METHODS After the oral administration of THC (120 mg/kg) for 5 consecutive days, a mouse model of sepsis was established via intraperitoneal lipopolysaccharide (LPS, 10 mg/kg) injection. Following this, cardiac function was assessed, pathological section staining was performed, and inflammatory markers were detected. RESULTS Myocardial systolic function was severely compromised in parallel with the accumulation of reactive oxygen species and enhanced cardiomyocyte apoptosis in mice with sepsis. These adverse changes were markedly reversed in response to THC treatment in septic mice as well as in LPS-treated H9c2 cells. Mechanistically, THC inhibited the release of pro-inflammatory cytokines, including tumor necrosis factor alpha, interleukin (IL)-1β, and IL-6, by upregulating mitogen-activated protein kinase phosphatase 1, to block the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated protein kinase (ERK). Additionally, THC enhanced the levels of antioxidant proteins, including nuclear factor-erythroid 2-related factor 2, superoxide dismutase 2, and NAD(P)H quinone oxidoreductase 1, while decreasing gp91phox expression. Furthermore, upon THC treatment, Bcl-2 expression was significantly increased, along with a decline in Bax and cleaved caspase-3 expression, which reduced cardiomyocyte loss. CONCLUSION Our findings indicate that THC exhibited protective potential against septic cardiomyopathy by reducing oxidative stress and inflammation through the regulation of JNK/ERK signaling. The findings of this study provide a basis for the further evaluation of THC as a therapeutic agent against septic cardiomyopathy.
Collapse
Affiliation(s)
- Hanzhao Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Liyun Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Hao Jia
- Department of Chemistry, Sacred Heart University, Fairfield, CT 06825, United States
| | - Lu Xu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Yu Cao
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Mengen Zhai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Kaifeng Li
- Basic Medical Teaching Experiment Center, Basic Medical College, The Air Force Medical University Xi'an, Shaanxi 710032, China
| | - Lin Xia
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning 110015, China
| | - Liqing Jiang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Xiang Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Yenong Zhou
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Jincheng Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Shiqiang Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Weixun Duan
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
21
|
Lecronier M, Jung B, Molinari N, Pinot J, Similowski T, Jaber S, Demoule A, Dres M. Severe but reversible impaired diaphragm function in septic mechanically ventilated patients. Ann Intensive Care 2022; 12:34. [PMID: 35403916 PMCID: PMC9001790 DOI: 10.1186/s13613-022-01005-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Whether sepsis-associated diaphragm dysfunction may improve despite the exposure of mechanical ventilation in critically ill patients is unclear. This study aims at describing the diaphragm function time course of septic and non-septic mechanically ventilated patients. Methods Secondary analysis of two prospective observational studies of mechanically ventilated patients in whom diaphragm function was assessed twice: within the 24 h after intubation and when patients were switched to pressure support mode, by measuring the endotracheal pressure in response to bilateral anterior magnetic phrenic nerve stimulation (Ptr,stim). Change in diaphragm function was expressed as the difference between Ptr,stim measured under pressure support mode and Ptr,stim measured within the 24 h after intubation. Sepsis was defined according to the Sepsis-3 international guidelines upon inclusion. In a sub-group of patients, the right hemidiaphragm thickness was measured by ultrasound. Results Ninety-two patients were enrolled in the study. Sepsis upon intubation was present in 51 (55%) patients. In septic patients, primary reason for ventilation was acute respiratory failure related to pneumonia (37/51; 73%). In non-septic patients, main reasons for ventilation were acute respiratory failure not related to pneumonia (16/41; 39%), coma (13/41; 32%) and cardiac arrest (6/41; 15%). Ptr,stim within 24 h after intubation was lower in septic patients as compared to non-septic patients: 6.3 (4.9–8.7) cmH2O vs. 9.8 (7.0–14.2) cmH2O (p = 0.004), respectively. The median (interquartile) duration of mechanical ventilation between first and second diaphragm evaluation was 4 (2–6) days in septic patients and 3 (2–4) days in non-septic patients (p = 0.073). Between first and second measurements, the change in Ptr,stim was + 19% (− 13–61) in septic patients and − 7% (− 40–12) in non-septic patients (p = 0.005). In the sub-group of patients with ultrasound measurements, end-expiratory diaphragm thickness decreased in both, septic and non-septic patients. The 28-day mortality was higher in patients with decrease or no change in diaphragm function. Conclusion Septic patients were associated with a more severe but reversible impaired diaphragm function as compared to non-septic patients. Increase in diaphragm function was associated with a better survival. Supplementary Information The online version contains supplementary material available at 10.1186/s13613-022-01005-9.
Collapse
Affiliation(s)
- Marie Lecronier
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France. .,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France.
| | - Boris Jung
- Département de Médecine Intensive - Réanimation, CHU Montpellier, Montpellier, France.,Laboratoire de Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046-CNRS UMR 9214, Université de Montpellier, Montpellier, France
| | - Nicolas Molinari
- Department of Medical Information, Hôpital Arnaud de Villeneuve, IMAG U5149, Université de Montpellier, Montpellier, France
| | - Jérôme Pinot
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Thomas Similowski
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France.,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France
| | - Samir Jaber
- Département de Médecine Intensive - Réanimation, CHU Montpellier, Montpellier, France.,Laboratoire de Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046-CNRS UMR 9214, Université de Montpellier, Montpellier, France
| | - Alexandre Demoule
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France.,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France
| | - Martin Dres
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France.,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France
| |
Collapse
|
22
|
Jahandideh F, Panahi S, Noble RMN, Gragasin FS, Khadaroo RG, Macala KF, Bourque SL. Characterization of Systemic and Regional Hemodynamics and Vascular Dysfunction in Mice with Fecal Induced Peritonitis. Biomedicines 2022; 10:biomedicines10020470. [PMID: 35203689 PMCID: PMC8962278 DOI: 10.3390/biomedicines10020470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 01/09/2023] Open
Abstract
Sepsis is associated with circulatory dysfunction contributing to disturbed blood flow and organ injury. Decreased organ perfusion in sepsis is attributed, in part, to the loss of vasoregulatory mechanisms. Identifying which vascular beds are most susceptible to dysfunction is important for monitoring the recovery of organ function and guiding interventions. This study aimed to investigate the development of vascular dysfunction as sepsis progressed to septic shock. Anesthetized C57Bl/6 mice were instrumented with a fiberoptic pressure sensor in the carotid artery for blood pressure measurements. In subgroups of mice, regional blood flow measurements were taken by positioning a perivascular flow probe around either the left carotid, left renal, or superior mesenteric arteries. Hemodynamic parameters and their responsiveness to bolus doses of vasoactive drugs were recorded prior to and continuously after injection of fecal slurry (1.3 mg/g body weight) for 4 h. Fecal slurry-induced peritonitis reduced mean arterial pressure (62.7 ± 2.4 mmHg vs. 37.5 ± 3.2 mmHg in vehicle and septic mice, respectively), impaired cardiac function, and eventually reduced organ blood flow (71.9%, 66.8%, and 65.1% in the superior mesenteric, renal, and carotid arteries, respectively). The mesenteric vasculature exhibited dysregulation before the renal and carotid arteries, and this underlying dysfunction preceded the blood pressure decline and impaired organ blood flow.
Collapse
Affiliation(s)
- Forough Jahandideh
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.J.); (S.P.); (F.S.G.); (K.F.M.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada;
| | - Sareh Panahi
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.J.); (S.P.); (F.S.G.); (K.F.M.)
| | - Ronan M. N. Noble
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Ferrante S. Gragasin
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.J.); (S.P.); (F.S.G.); (K.F.M.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada;
| | - Rachel G. Khadaroo
- Department of Critical Care Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada;
- Department of Surgery, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Kimberly F. Macala
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.J.); (S.P.); (F.S.G.); (K.F.M.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada;
- Department of Critical Care Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada;
| | - Stephane L. Bourque
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.J.); (S.P.); (F.S.G.); (K.F.M.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Correspondence: ; Tel.: +1-780-492-6000
| |
Collapse
|
23
|
Liu L, Li Y, Chen Q. The Emerging Role of FUNDC1-Mediated Mitophagy in Cardiovascular Diseases. Front Physiol 2022; 12:807654. [PMID: 34975548 PMCID: PMC8718682 DOI: 10.3389/fphys.2021.807654] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/19/2021] [Indexed: 01/27/2023] Open
Abstract
Mitochondria are highly dynamic organelles and play essential role in ATP synthase, ROS production, innate immunity, and apoptosis. Mitochondria quality control is critical for maintaining the cellular function in response to cellular stress, growth, and differentiation Signals. Damaged or unwanted mitochondria are selectively removed by mitophagy, which is a crucial determinant of cell viability. Mitochondria-associated Endoplasmic Reticulum Membranes (MAMs) are the cellular structures that connect the ER and mitochondria and are involved in calcium signaling, lipid transfer, mitochondrial dynamic, and mitophagy. Abnormal mitochondrial quality induced by mitophagy impairment and MAMs dysfunction is associated with many diseases, including cardiovascular diseases (CVDs), metabolic syndrome, and neurodegenerative diseases. As a mitophagy receptor, FUNDC1 plays pivotal role in mitochondrial quality control through regulation of mitophagy and MAMs and is closely related to the occurrence of several types of CVDs. This review covers the regulation mechanism of FUNDC1-mediated mitophagy and MAMs formation, with a particular focus on its role in CVDs.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yimei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Quan Chen
- Interdisciplinary Center of Cell Response, State key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
24
|
Asci H, Ozmen O, Erzurumlu Y, Sofu A, Icten P, Kaynak M. Agomelatine protects heart and aorta against lipopolysaccharide-induced cardiovascular toxicity via inhibition of NF-kβ phosphorylation. Drug Chem Toxicol 2022; 45:133-142. [PMID: 31514555 DOI: 10.1080/01480545.2019.1663209] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022]
Abstract
The aim of this study was to investigate the possible ameliorating effects of agomelatine (AGO) on lipopolysaccharide (LPS)-induced endothelial and cardiac damage. Twenty-four female Wistar Albino rats divided into 3 groups as follows: Control, LPS and LPS + AGO. Total oxidant status (TOS), total antioxidant status (TAS), nuclear factor kappa beta (NF-kβ)/p65, p-NF-kβ, full caspase-8 (Cas-8) and cleaved cas-8 levels were measured in cardiac tissues and creatine kinase MB (CKMB), aspartate aminotransferase (AST), lactate dehydrogenase (LDH) levels in blood biochemically. In addition; cas-8, sirtuin-1 (SIRT-1), interleukin-4 (IL-4), interleukin-10 (IL-10), haptoglobin measured histopathologically in cardiac and aortic tissues. The levels of CKMB, AST, LDH and TOS were increased and TAS were decreased in the LPS group. In Western blot analyses NF-kβ/p65, p-NF-kβ/p65, full and cleaved cas-8 protein levels increased in cardiac tissues of LPS group. In histopathological and immunohistochemical evaluation of the heart sections; hyperemia, micro-hemorrhages and inflammatory cell infiltrations, increase of cas-8, haptoglobin, IL-4 and IL-10 and decrease of SIRT-1 levels were observed in cardiac and endothelial tissues of LPS groups. AGO treatment reversed all these parameters. It was shown that LPS-induced inflammation, oxidative stress and apoptosis via increasing of NF-kβ/p65 signaling, decreasing of SIRT-1 levels and increase of cas-8 levels in heart and endothelial tissues respectively. AGO corrected all these parameters by its antioxidant, antiinflammatory and antiapoptotic activities.
Collapse
Affiliation(s)
- H Asci
- Department of Pharmacology, Faculty of Medicine/Medicine, Medical Device and Dermocosmetic Research and Application Laboratory (IDAL), Suleyman Demirel University, Isparta, Turkey
| | - O Ozmen
- Department of Pathology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Y Erzurumlu
- Department of Biochemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
| | - A Sofu
- Department of Bioengineering, Faculty of Engineering, Suleyman Demirel University, Isparta, Turkey
| | - P Icten
- Department of Pharmacology, Faculty of Medicine/Medicine, Medical Device and Dermocosmetic Research and Application Laboratory (IDAL), Suleyman Demirel University, Isparta, Turkey
| | - M Kaynak
- Department of Pharmacology, Faculty of Medicine/Medicine, Medical Device and Dermocosmetic Research and Application Laboratory (IDAL), Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
25
|
Liu S, Chong W. Roles of LncRNAs in Regulating Mitochondrial Dysfunction in Septic Cardiomyopathy. Front Immunol 2021; 12:802085. [PMID: 34899764 PMCID: PMC8652231 DOI: 10.3389/fimmu.2021.802085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/10/2021] [Indexed: 01/20/2023] Open
Abstract
Sepsis is an abnormal systemic inflammatory response of the host immune system to infection and can lead to fatal multiorgan dysfunction syndrome. Epidemiological studies have shown that approximately 10-70% of sepsis cases can lead to septic cardiomyopathy. Since the pathogenesis of septic cardiomyopathy is not clear, it is difficult for medical doctors to treat the disease. Therefore, finding effective interventions to prevent and reduce myocardial damage in septic cardiomyopathy is clinically significant. Epigenetics is the study of stable genetic phenotype inheritance that does not involve changing gene sequences. Epigenetic inheritance is affected by both gene and environmental regulation. Epigenetic studies focus on the modification and influence of chromatin structure, mainly including chromatin remodelling, DNA methylation, histone modification and noncoding RNA (ncRNA)-related mechanisms. Recently, long ncRNA (lncRNA)-related mechanisms have been the focus of epigenetic studies. LncRNAs are expected to become important targets to prevent, diagnose and treat human diseases. As the energy metabolism centre of cells, mitochondria are important targets in septic cardiomyopathy. Intervention measures to prevent and treat mitochondrial damage are of great significance for improving the prognosis of septic cardiomyopathy. LncRNAs play important roles in life activities. Recently, studies have focused on the involvement of lncRNAs in regulating mitochondrial dysfunction. However, few studies have revealed the involvement of lncRNAs in regulating mitochondrial dysfunction in septic cardiomyopathy. In this article, we briefly review recent research in this area.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Chong
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Xu Z, Lin X, Zhu J, Zhu Z. Long noncoding RNAs Colorectal Neoplasia Differentially Expressed and taurine-upregulated gene 1 are downregulated in sepsis and positively regulate each other to suppress the apoptosis of cardiomyocytes. Bioengineered 2021; 12:11369-11375. [PMID: 34872438 PMCID: PMC8810183 DOI: 10.1080/21655979.2021.2008658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) Colorectal Neoplasia Differentially Expressed (CRNDE) and taurine-upregulated gene 1 (TUG1) play similar roles in sepsis, indicating the existence of the crosstalk between them. Sepsis is a major cause of injuries in heart, which are related to high mortality rates. This study was therefore carried out to analyze the potential crosstalk between CRNDE and TUG1 in sepsis, with a focus on sepsis-induced cell apoptosis in heart. Expression of CRNDE and TUG1 was analyzed with RT-qPCR. Correlations between them were analyzed by Pearson’s correlation coefficient. CRNDE and TUG1 were overexpressed in cardiomyocytes to determine the relationship between them. The roles of CRNDE and TUG1 in regulating the apoptosis of cardiomyocytes were explored by cell apoptosis assay. We found that both CRNDE and TUG1 were downregulated in sepsis. In cardiomyocytes, LPS treatment resulted in the downregulation of CRNDE and TUG1. Overexpression of CRNDE and TUG1 in cardiomyocytes increased the expression levels of each other. Under lipopolysaccharide (LPS) treatment, decreased apoptosis rates of cardiomyocytes were observed after CRNDE and TUG1 overexpression. CRNDE and TUG1 co-overexpression showed a stronger effect. In conclusion, CRNDE and TUG1 are downregulated in sepsis and they positively regulate each other to suppress the apoptosis of cardiomyocytes.
Collapse
Affiliation(s)
- Zhenwei Xu
- Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou City, Fujian Province, PR. China
| | - Xingyu Lin
- Department of Emergency, Fujian Medical University Union Hospital, Fuzhou City, Fujian Province, PR China
| | - Jingfa Zhu
- Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou City, Fujian Province, PR. China
| | - Zhixia Zhu
- Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou City, Fujian Province, PR. China
| |
Collapse
|
27
|
Chen M, Li J, Wang J, Le Y, Liu C. SMYD1 alleviates septic myocardial injury by inhibiting endoplasmic reticulum stress. Biosci Biotechnol Biochem 2021; 85:2383-2391. [PMID: 34601561 DOI: 10.1093/bbb/zbab167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/21/2021] [Indexed: 11/14/2022]
Abstract
Sepsis-induced cardiomyopathy (SIC) is a major complication of sepsis. SET and MYND domain containing 1 (SMYD1) has central importance in heart development, and its role in SIC has not been identified. Herein, we found that the expression of SMYD1 was downregulated in myocardial tissues of SIC patients (from GEO database: GSE79962) and lipopolysaccharide (LPS)-induced SIC rats, and LPS-induced H9c2 cardiomyocytes. We used LPS-stimulated H9c2 cells that mimic sepsis in vitro to explore the function of SMYD1 in SIC. MTT assay, LDH and CK-MB release assay, flow cytometry, and ELISA assay showed that SMYD1 overexpression enhanced cell viability, alleviated cell injury, impeded apoptosis, and reduced the level of proinflammatory factors and NF-κB activation under the condition of LPS stimulation. Moreover, SMYD1 exerted protective effect on H9c2 cells stimulated with LPS through relieving endoplasmic reticulum (ER) stress. In conclusion, overexpression of SMYD1 alleviates cardiac injury through relieving ER stress during sepsis.
Collapse
Affiliation(s)
- Meixue Chen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Pediatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jing Li
- Department of Pediatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jinfeng Wang
- Department of Pediatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuan Le
- Department of Pediatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chunfeng Liu
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
28
|
Liang YW, Zhu YF, Zhang R, Zhang M, Ye XL, Wei JR. Incidence, prognosis, and risk factors of sepsis-induced cardiomyopathy. World J Clin Cases 2021; 9:9452-9468. [PMID: 34877280 PMCID: PMC8610866 DOI: 10.12998/wjcc.v9.i31.9452] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/20/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND At present, large-scale studies on the clinical characteristics of sepsis-induced cardiomyopathy (SIC) are lacking.
AIM To investigate the clinical characteristics of SIC.
METHODS Based on the analysis of the MIMIC-III public database, we performed a large-scale retrospective study involving sepsis patients who were admitted to the intensive care unit (ICU) and had no concomitant cardiac disease. We used propensity score matching analysis and multivariate logistic regression to ensure the robustness of the results. The primary outcome was hospital mortality, and the secondary outcomes included the number of patients who received mechanical ventilation or renal replacement therapy during their hospital stay, the number of patients administered with vasopressors, the length of ICU stay, and the length of hospital stay.
RESULTS In the present study, after screening 38605 patients, 3530 patients with sepsis were included. A total of 997 patients met the SIC diagnostic criteria, and the incidence of SIC was 28.20% (95% confidence interval [CI]: 26.80%-29.70%). Compared to patients in the non-SIC group, patients in the SIC group were of older age and had a higher Simplified Acute Physiology Score (SAPS)-I score, SAPS-II score, and Elixhauser comorbidity index (ECI). A total of 367 (36.8%) of 997 patients in the SIC group and 818 (32.3%) of 2533 patients in the non-SIC group died in the hospital, which resulted in a significant between-group difference (odds ratios = 1.22, 95%CI: 1.05-1.42; P = 0.011). For the secondary outcomes, more patients in the SIC group received mechanical ventilation and vasopressors. Multivariate logistic regression analysis showed that age, male sex, ECI, hemoglobin level, diabetes, and mechanical ventilation use on the first day of ICU admission were risk factors for SIC.
CONCLUSION Compared with non-SIC patients, hospital mortality is higher in SIC patients.
Collapse
Affiliation(s)
- Yan-Wen Liang
- Department of Intensive Care Unit, Guangzhou Red Cross Hospital, Guangzhou 510220, Guangdong Province, China
| | - You-Feng Zhu
- Department of Intensive Care Unit, Guangzhou Red Cross Hospital, Guangzhou 510220, Guangdong Province, China
| | - Rui Zhang
- Department of Intensive Care Unit, Guangzhou Red Cross Hospital, Guangzhou 510220, Guangdong Province, China
| | - Min Zhang
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, Qingdao 270000, Shandong Province, China
| | - Xiao-Ling Ye
- Department of Intensive Care Unit, Guangzhou Red Cross Hospital, Guangzhou 510220, Guangdong Province, China
| | - Jian-Rui Wei
- Guangzhou Women and Children’s Medical Center, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
29
|
Cioccari L, Jakob SM, Takala J. Should Vasopressors Be Started Early in Septic Shock? Semin Respir Crit Care Med 2021; 42:683-688. [PMID: 34544185 DOI: 10.1055/s-0041-1733897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Sepsis can influence blood volume, its distribution, vascular tone, and cardiac function. Persistent hypotension or the need for vasopressors after volume resuscitation is part of the definition of septic shock. Since increased positive fluid balance has been associated with increased morbidity and mortality in sepsis, timing of vasopressors in the treatment of septic shock seems crucial. However, conclusive evidence on timing and sequence of interventions with the goal to restore tissue perfusion is lacking. The aim of this narrative review is to depict the pathophysiology of hypotension in sepsis, evaluate how common interventions to treat hypotension interfere with physiology, and to give a resume of the results from clinical studies focusing on targets and timing of vasopressor in sepsis. The majority of studies comparing early versus late administration of vasopressors in septic shock are rather small, single-center, and retrospective. The range of "early" is between 1 and 12 hours. The available studies suggest a mean arterial pressure of 60 to 65 mm Hg as a threshold for increased risk of morbidity and mortality, whereas higher blood pressure targets do not seem to add further benefits. The data, albeit mostly from observational studies, speak for combining vasopressors with fluids rather "early" in the treatment of septic shock (within a 0-3-hour window). Nevertheless, the optimal resuscitation strategy should take into account the source of infection, the pathophysiology, the time and clinical course preceding the diagnosis of sepsis, and also comorbidities and sepsis-induced organ dysfunction.
Collapse
Affiliation(s)
- Luca Cioccari
- Department of Intensive Care Medicine, University of Bern, Bern University Hospital, Bern, Switzerland
| | - Stephan M Jakob
- Department of Intensive Care Medicine, University of Bern, Bern University Hospital, Bern, Switzerland
| | - Jukka Takala
- Department of Intensive Care Medicine, University of Bern, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
30
|
Clinical impact of visually assessed right ventricular dysfunction in patients with septic shock. Sci Rep 2021; 11:18823. [PMID: 34552188 PMCID: PMC8458318 DOI: 10.1038/s41598-021-98397-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022] Open
Abstract
We retrospectively analyzed data from the Medical Information Mart for Intensive Care-III critical care database to determine whether visually-assessed right ventricular (RV) dysfunction was associated with clinical outcomes in septic shock patients. Associations between visually-assessed RV dysfunction by echocardiography and in-hospital mortality, lethal arrhythmia, and hemodynamic indicators to determine the prognostic value of RV dysfunction in patients with septic shock were analyzed. Propensity score analysis showed RV dysfunction was associated with increased risk of in-hospital death in patients with septic shock (adjusted odds ratio [OR] 2.15; 95% confidence interval [CI] 1.99–2.32; P < 0.001). In multivariate logistic regression analysis, RV dysfunction was associated with in-hospital death (OR 2.19; 95% CI 1.91–2.53; P < 0.001), lethal arrhythmia (OR 2.19; 95% CI 1.34–3.57; P < 0.001), and tendency for increased blood lactate levels (OR 1.31; 95% CI 1.14–1.50; P < 0.001) independent of left ventricular (LV) dysfunction. RV dysfunction was associated with lower cardiac output, pulmonary artery pressure index, and RV stroke work index. In patients with septic shock, visually-assessed RV dysfunction was associated with in-hospital mortality, lethal arrhythmia, and circulatory insufficiency independent of LV dysfunction. Visual assessment of RV dysfunction using echocardiography might help to identify the short-term prognosis of patients with septic shock by reflecting hemodynamic status.
Collapse
|
31
|
Song B, Wang XX, Yang HY, Kong LT, Sun HY. MiR-141 attenuates sepsis-induced cardiomyopathy by targeting DAPK1. Hum Exp Toxicol 2021; 40:S137-S149. [PMID: 34289745 DOI: 10.1177/09603271211033768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To discuss the possible effects of microRNA-141 (miR-141) in sepsis-induced cardiomyopathy (SIC) via targeting death-associated protein kinase 1 (DAPK1). METHODS An SIC mouse model was constructed by abdominal injection of lipopolysaccharide (LPS) and divided into control, LPS, LPS + pre-miR-141, and LPS + anti-miR-141 groups. Hemodynamic indicators and heart function indexes of mice were detected. ELISA was used to determine the serum levels of inflammatory cytokines, while TUNEL staining to observe the apoptosis of myocardial cells of mice, as well as qRT-PCR and Western blotting to clarify the expression of miR-141 and DAPK1. Lastly, in vitro experiment was also conducted on the primary neonatal rat ventricular cardiomyocytes (NRVCMs) to validate the results. RESULTS Mice in the LPS group, as compared to the control group, had lower left ventricular ejection fraction, left ventricular fractional shortening, left ventricular systolic pressure, and ±dp/dt, but a higher left ventricular end-diastolic pressure, while the serum expression of IL-1β, IL-6, TNF-α, and cTn-T was up-regulated evidently with the increased apoptotic index of myocardial tissues. However, miR-141 and Bcl-2/Bax were down-regulated with elevated DAPK1 and cleaved caspase-3. The above changes were ameliorated in mice from the LPS + pre-miR-141 group relative to the LPS group, while those in the LPS + anti-miR-141 group were further deteriorated. In vitro experiment showed that miR-141 overexpression could reduce the apoptosis of LPS-induced NRVCMs and the levels of inflammatory cytokines with the increased cell viability. CONCLUSION MiR-141 could decrease inflammatory response and reduce myocardial cell apoptosis by targeting DAPK1, thereby playing the promising protective role in SIC.
Collapse
Affiliation(s)
- Bo Song
- Department of Emergency, 519688YanTaiShan Hospital, YanTai, China
| | - Xin-Xiang Wang
- Yantai Chefoo Area Directly Subordinate Organ Hospital, Yantai, China
| | - Hai-Yan Yang
- Department of Emergency, 519688YanTaiShan Hospital, YanTai, China
| | - Ling-Ting Kong
- Department of Emergency, 519688YanTaiShan Hospital, YanTai, China
| | - Hong-Yan Sun
- Department of Endocrinology, 519688YanTaiShan Hospital, YanTai, China
| |
Collapse
|
32
|
Wang R, Xu Y, Fang Y, Wang C, Xue Y, Wang F, Cheng J, Ren H, Wang J, Guo W, Liu L, Zhang M. Pathogenetic mechanisms of septic cardiomyopathy. J Cell Physiol 2021; 237:49-58. [PMID: 34278573 DOI: 10.1002/jcp.30527] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/17/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022]
Abstract
Sepsis is a serious complication after infection, whose further development may lead to multiple organ dysfunction syndrome and so on. It is an important cause of death in critically ill patients who suffered an infection. Sepsis cardiomyopathy is a common complication that exacerbates the prognosis of patients. At present, though the pathogenesis of sepsis cardiomyopathy is not completely clear, in-depth study of the pathogenesis of sepsis cardiomyopathy and the discovery of its potential therapeutic targets may decrease the mortality of sepsis patients and bring clinical benefits. This article reviews mitochondrial dysfunction, mitophagy, oxidation stress, and other mechanisms in sepsis cardiomyopathy.
Collapse
Affiliation(s)
- Runze Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Hematology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chiyao Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yugang Xue
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Fangfang Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jin Cheng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - He Ren
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jie Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wangang Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Li Liu
- Department of Hematology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
33
|
Hai PD, Binh NT, Tot NH, Hung HM, Hoa LTV, Hien NVQ, Son PN. Diagnostic Value of High-Sensitivity Troponin T for Subclinical Left Ventricular Systolic Dysfunction in Patients with Sepsis. Cardiol Res Pract 2021; 2021:8897738. [PMID: 33981455 PMCID: PMC8088348 DOI: 10.1155/2021/8897738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Left ventricular systolic dysfunction (LVSD) is common in sepsis. Speckle-tracking echocardiography (STE) is a useful emerging tool for evaluating the intrinsic left ventricular systolic function. High-sensitivity cardiac troponin T (hs-cTnT) is the most sensitive biomarker of myocardial injury. However, there are limited data regarding the association between hs-cTnT level and left ventricular systolic dysfunction based on STE in septic patients. We performed this prospective study to evaluate the diagnostic value of hs-cTnT level for subclinical left ventricular systolic dysfunction measured by STE in septic patients according to the sepsis-3 definition. METHODS Patients with sepsis based on sepsis-3 definition admitted to the intensive care unit were prospectively performed STE and hs-cTnT level within 24 hours after the onset of sepsis. Baseline clinical and echocardiographic variables were collected. Left ventricular systolic dysfunction was defined as a global longitudinal strain of ≥-15%. RESULTS During a 19-month period, 116 patients were enrolled in the study. The elevated hs-cTnT level was seen in 86.2% of septic patients, and 43.1% of patients had LVSD on STE. The median hs-cTnT level and the proportion of elevated hs-cTnT level (>14 ng/L) were significantly higher in patients with LVSD than in patients without LVSD. The area under the ROC curves of hs-cTnT to detect LVSD was 0.73 (P < 0.001). In the multivariate analysis, hs-cTnT (HR, 1.002; 95% CI, 1.000 to 1.004; P = 0.025) and septic shock (HR, 7.6; 95% CI, 2.25 to 25.76; P = 0.001) were independent predictors of LVSD. CONCLUSION Our study indicated that the serum hs-cTnT level might be a useful biomarker for detecting LVSD in septic patients.
Collapse
Affiliation(s)
- Pham Dang Hai
- Intensive Care Unit, 108 Military Central Hospital, Hanoi, Vietnam
| | | | - Nguyen Hong Tot
- Intensive Care Unit, 108 Military Central Hospital, Hanoi, Vietnam
| | - Ha Manh Hung
- Intensive Care Unit, 108 Military Central Hospital, Hanoi, Vietnam
| | | | - Nguyen Viet Quang Hien
- Department of Anesthesiology and Critical Care Medicine, Hue Central Hospital, Hue, Vietnam
| | - Pham Nguyen Son
- Department of Cardiology, 108 Military Central Hospital, Hanoi, Vietnam
| |
Collapse
|
34
|
Huang Q, Liu DH, Chen CF, Han Y, Huang ZQ, Zhang JW, Zeng XM. Pgc-1α Promotes Phosphorylation, Inflammation, and Apoptosis in H9c2 Cells During the Early Stage of Lipopolysaccharide Induction. Inflammation 2021; 44:1771-1781. [PMID: 33847873 DOI: 10.1007/s10753-021-01453-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/05/2021] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
Cardiac dysfunction in severe sepsis is associated with increased mortality. However, the molecular mechanisms underlying septic heart dysfunction remain unclear. Expression of peroxisome proliferator-activated receptor-γ coactivator 1α (Pgc-1α), concentrations of inflammatory factors, and activation of the nuclear factor kappa-B (NF-κB) signaling pathway were examined in H9c2 cells after a 24-h lipopolysaccharide (LPS) stimulation period using qPCR, enzyme-linked immunosorbent assays (ELISAs), and western blots (WBs), respectively. Pgc-1α was overexpressed and suppressed in cells using a lentivirus vector and siRNA, respectively. The effects of Pgc-1α dysfunction on the release of inflammatory factors and apoptosis were analyzed. Pgc-1α expression was increased after LPS induction for 0.5 h and returned to the pre-induction level at 2 h. Levels of IL-1β, IL-6, and TNF-α increase after LPS induction for 0.5 h and accumulated in the culture supernatants over time. The WBs revealed the highest Pgc-1α and phospho (p)-p65 protein levels after LPS induction for 0.5 h, followed by a decrease; moreover, the cleaved-caspase-3 level increased after LPS induction for 0.5 h and increased gradually thereafter. A functional analysis of Pgc-1α revealed that overexpression of this protein enhanced LPS-induced inflammatory factors and p-p65 levels and inhibited apoptosis during the early stage after LPS induction (0.5 and 4 h). In contrast, the inhibition of Pgc-1α expression inhibited the LPS expression-associated increases in inflammatory factors and p-p65 and promoted apoptosis. Pgc-1α promoted LPS-induced p65 phosphorylation and inflammatory factor release while inhibiting apoptosis.
Collapse
Affiliation(s)
- Qun Huang
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China
| | - De-Hong Liu
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China.
| | - Chang-Feng Chen
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China
| | - Yong Han
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China
| | - Zhi-Qiang Huang
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China
| | - Ji-Wen Zhang
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China
| | - Xiao-Mei Zeng
- Emergency Department, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, No.3002 Sungang West Road, Shenzhen, 518035, Guangdong, People's Republic of China
| |
Collapse
|
35
|
Anti-Interleukin-16 Neutralizing Antibody Treatment Alleviates Sepsis-Induced Cardiac Injury and Dysfunction via the Nuclear Factor Erythroid-2 Related Factor 2 Pathway in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6616422. [PMID: 33628366 PMCID: PMC7896865 DOI: 10.1155/2021/6616422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 11/17/2022]
Abstract
Several interleukin (IL) members have been reported to participate in sepsis. In this study, the effects of IL-16 on sepsis-induced cardiac injury and dysfunction were examined, and the related mechanisms were detected. IL-16 expression in septic mice was first measured, and the results showed that both cardiac and serum IL-16 expression levels were increased in mice with sepsis induced by LPS or cecal ligation and puncture (CLP) compared with control mice. Then, IL-16 was neutralized, and the effects on lipopolysaccharide- (LPS-) induced cardiac injury were detected. The results showed that an anti-IL-16 neutralizing antibody (nAb) significantly reduced mortality and increased serum lactate dehydrogenase (LDH), creatine kinase myocardial bound (CK-MB), and cardiac troponin T (cTnT) levels while improving cardiac function in mice with LPS-induced sepsis. Neutralization of IL-16 also increased the activation of antioxidant pathways and the expression of antioxidant factors in septic mice while decreasing the activation of prooxidant pathways and the expression of prooxidants. Treatment with the anti-IL-16 nAb increased mitochondrial apoptosis-inducing factor (AIF) expression, decreased nuclear AIF and cleaved poly-ADP-ribose polymerase (PARP) expression, and decreased TUNEL-positive cell percentages in LPS-treated mice. Additionally, treatment with CPUY192018, the nuclear factor erythroid-2 related factor 2 (Nrf2) pathway, significantly increased mortality and reversed the above effects in mice treated with LPS and the anti-IL-16 nAb. Our results showed that the anti-IL-16 nAb regulates oxidative stress through the Nrf2 pathway and participates in the regulation of cardiac injury in septic mice. Neutralization of IL-16 may be a beneficial strategy for the prevention of cardiac injury and dysfunction in sepsis patients.
Collapse
|
36
|
Yang X, Sun J, Sun F, Yao Y, Tian T, Zhou J, Shen W, Lu M, Lei M. TRIM31 promotes apoptosis via TAK1-mediated activation of NF-κB signaling in sepsis-induced myocardial dysfunction. Cell Cycle 2020; 19:2685-2700. [PMID: 33016203 DOI: 10.1080/15384101.2020.1826235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Sepsis is a major condition caused by an overwhelming inflammatory response to an infection. Sepsis-induced myocardial dysfunction (SIMD) is a common complication in septic patients and a major predictor of morbidity and mortality. Here, we investigated the role of tripartite motif 31 (TRIM31) protein in sepsis progression in vitro and in vivo. Quantitative real-time PCR and western blot were used to detect the expression levels of relative genes and proteins. Cell proliferation and apoptosis were evaluated to determine cell viability. H&E and IHC staining were performed to examine morphological and pathological changes in mice. ELISA assay was used to detect inflammatory factors. TRIM31 was upregulated in septic patients compared with normal people. TRIM31 depletion reduced LPS-induced apoptosis whereas TRIM31 overexpression-elevated LPS-induced apoptosis. Furthermore, TRIM31 interacted with and ubiquitinated transforming growth factor-β-activated kinase-1 (TAK1), resulting in TAK1 activation and subsequent induction of NF-κB signaling. Of note, Trim31 depletion or blockade by PDTC treatment inhibited LPS-induced apoptosis in vivo. In conclusion, TRIM31 played an important role in SIMD by activating TAK1-mediated NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaofang Yang
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Jingjing Sun
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - FangYuan Sun
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Yulong Yao
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Tianning Tian
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Jiayi Zhou
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Weihong Shen
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Ming Lu
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| | - Ming Lei
- Intensive Care Department of Shanghai Seventh People's Hospital , Shanghai, China
| |
Collapse
|
37
|
Tarasiuk E, Bonda TA, Dziemidowicz M, Winnicka MM, Bernaczyk P, Kamiński KA. The effect of interleukin 6 deficiency on myocardial signal transduction pathways activation induced by bacterial lipopolysaccharide in young and old mice. Adv Med Sci 2020; 65:386-393. [PMID: 32693349 DOI: 10.1016/j.advms.2020.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/13/2020] [Accepted: 06/20/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE Exaggerated release of proinflammatory mediators during sepsis contributes to inadequate vasodilatation and depressed myocardial contractility, which lead to development of shock and circulatory collapse. The aim of the study was to evaluate the effect of IL-6 and aging on activation of intracellular signaling pathways in the myocardium induced by bacterial lipopolysaccharide (LPS) administration. MATERIAL/METHODS LPS was injected intraperitoneally to male 3- and 24-month old mice with systemic IL-6 gene knock-out (IL-6KO) and the reference strain (WT). LPS was given intraperitoneally in single low (0.1 mg/kg) or high (10 mg/kg) dose, or in two doses (0.1 + 10 mg/kg) with 24-h delay. The expression and phosphorylation of STAT3, ERK1/2, Akt1/2/3 proteins in the left ventricular myocardium was evaluated after 24 h using Western blotting. RESULTS Low LPS dose induced higher STAT3 phosphorylation only in old IL-6KO mice, not affecting ERK1/2 and Akt1/2/3 phosphorylation in any group. High LPS dose upregulated STAT3 phosphorylation similarly in all groups, reduced ERK1/2 expression in young WT mice and upregulated Akt1/2/3 expression and phosphorylation in young IL-6KO mice. Pretreatment with low LPS dose attenuated phosphorylation of STAT3 in both old groups and phosphorylation of Akt1/2/3 in young IL-6KO group. Two-dose approach also significantly potentiated ERK1/2 phosphorylation in both old groups. CONCLUSIONS Obtained results show that IL-6 deficiency alters the activity of intracellular signaling pathways: JAK/STAT in old and Akt in young LPS-treated mice. This may indicate that lack of IL-6 attenuates Akt-related cytoprotective effect of pretreatment with low LPS dose in young but not in aged animals.
Collapse
Affiliation(s)
- Ewa Tarasiuk
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Tomasz A Bonda
- Department of General and Experimental Pathology, Medical University of Bialystok, Bialystok, Poland
| | - Magdalena Dziemidowicz
- Department of General and Experimental Pathology, Medical University of Bialystok, Bialystok, Poland
| | - Maria M Winnicka
- Department of General and Experimental Pathology, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Bernaczyk
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Karol A Kamiński
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland; Department of Population Medicine and Civilization Diseases Prevention, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
38
|
Yang Y, Zhu Y, Xiao J, Tian Y, Ma M, Li X, Li L, Zhang P, Li M, Wang J, Jin S. Maresin conjugates in tissue regeneration 1 prevents lipopolysaccharide-induced cardiac dysfunction through improvement of mitochondrial biogenesis and function. Biochem Pharmacol 2020; 177:114005. [DOI: 10.1016/j.bcp.2020.114005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022]
|
39
|
Jiang W, Li W, Hu X, Hu R, Li B, Lan L. CTRP1 prevents sepsis-induced cardiomyopathy via Sirt1-dependent pathways. Free Radic Biol Med 2020; 152:810-820. [PMID: 31991227 DOI: 10.1016/j.freeradbiomed.2020.01.178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/09/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
C1q/tumor necrosis factor-related protein 1 (CTRP1) has recently been identified as a key regulator of cardio-metabolic diseases. It has been reported that CTRP1 could inhibit the hypertrophic response in mice. However, the effect of CTRP1 on sepsis-induced cardiomyopathy remains completely unknown. Cardiomyocyte-specific CTRP1 overexpression was achieved using an adeno associated virus system in mice. CTRP1 deficiency mice were also subjected to lipopolysaccharide (LPS) injection. We found that CTRP1 overexpression improved survival rate and cardiac function, and suppressed myocardial inflammation, oxidative damage and apoptosis without affecting metabolic disturbance in LPS-treated mice. CTRP1 depletion further decreased survival rate and cardiac function, and promoting myocardial inflammation, oxidative damage and apoptosis in sepsis mice. In addition, we showed that CTRP1 provided protection against LPS-induced cell injury in vitro. CTRP1 activated sirtuin 1 (Sirt1) signaling pathway, and Sirt1 inhibition or deficiency blocked CTRP1-mediated cardioprotective effects in vivo and in vitro. More importantly, our study found that recombinant human globular domain of CTRP1 infusion was also capable of blocking sepsis-induced cardiomyopathy in mice. In conclusion, CTRP1 improved survival rate and attenuated LPS-induced cardiac injury via activating Sirt1 signaling pathway.
Collapse
Affiliation(s)
- Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen Li
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Rui Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bowen Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Linhui Lan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| |
Collapse
|
40
|
Guo Z, Wang Y, Xie C, Hua G, Ge S, Li Y. Effects of respiratory rate on venous-to-arterial CO 2 tension difference in septic shock patients undergoing volume mechanical ventilation. Eur J Med Res 2020; 25:6. [PMID: 32183893 PMCID: PMC7076974 DOI: 10.1186/s40001-020-00402-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/05/2020] [Indexed: 11/18/2022] Open
Abstract
Background To explore the effects of the respiratory rate (RR) on the venous-to-arterial CO2 tension difference (gapCO2) in septic shock patients undergoing volume mechanical ventilation. Methods Adult patients with septic shock underwent volume mechanical ventilation between October 2015 and October 2016. RR was started at 10 breaths/min, and 2 breaths/min were added every 60 min until 16 breaths/min was reached. At every point, central venous and arterial blood gas measurements were obtained simultaneously. Results In this study, gapCO2 induced by hyperventilation significantly increased, while the central venous carbon dioxide pressure (PvCO2) and the partial pressure of CO2 (PaCO2) in arteries decreased. The decreasing trend of the PaCO2 was more obvious than that of the PvCO2. HCO3− and ctCO2 were markedly decreased, when the RR was increased (P < 0.05). Central venous oxygen saturation (ScvO2) had a decreasing trend between 14 (77.1 ± 8.3%) and 16 (75.2 ± 8.7%) breaths/min; however, the difference was not significant. Conclusions In septic patients undergoing ventilation, respiratory alkalosis induced by hyperventilation caused an increase in the gapCO2. Clinicians should cautiously interpret the gapCO2 in hemodynamically stable ventilated septic shock patients and its relationship with low cardiac output and inadequate perfusion.
Collapse
Affiliation(s)
- Zhixiang Guo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yapeng Wang
- Department of Cardiac Intensive Care Unit (CICU), The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Chao Xie
- Department of Critical Care Medicine, The 901st Hospital of the Joint Logistics Support Force of PLA, No. 424 Changjiang West Road, Hefei, 230000, Anhui, China
| | - Guofang Hua
- Department of Critical Care Medicine, The 901st Hospital of the Joint Logistics Support Force of PLA, No. 424 Changjiang West Road, Hefei, 230000, Anhui, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Yuedong Li
- Department of Critical Care Medicine, The 901st Hospital of the Joint Logistics Support Force of PLA, No. 424 Changjiang West Road, Hefei, 230000, Anhui, China.
| |
Collapse
|
41
|
Shin DG, Kang MK, Seo YB, Choi J, Choi SY, Choi S, Cho JR, Lee N. Factors associated with abnormal left ventricular ejection fraction (decreased or increased) in patients with sepsis in the intensive care unit. PLoS One 2020; 15:e0229563. [PMID: 32155161 PMCID: PMC7064219 DOI: 10.1371/journal.pone.0229563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/09/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Sepsis-induced cardiomyopathy (SIC) is known to show cardiac dysfunction in patients with sepsis. Both a decrease or an increase in ejection fraction (EF), an indicator of cardiac function, can occur. The purpose of this study was to identify factors associated with abnormal left ventricular (LV) function measured by EF in patients with sepsis in the intensive care unit (ICU). METHODS This was a retrospective study performed from November 2016 to December 2018. Three-hundred and sixty-six patients (mean age, 73 ± 13 years; 191 [52%] men) admitted to the ICU with sepsis were included. Patients were classified into three categories according to LV EF (group 1 -[EF<50%, n = 36], group 2 -[50≤EF<70%, n = 252], and group 3 -[EF≥70%, n = 78]). Echocardiographic assessment was performed within 48 hours of diagnosis of sepsis. We analyzed clinical factors including mortality, echocardiographic findings, and laboratory parameters. RESULTS Decreased LV EF occurred in 36 (10%) patients and hyper-dynamic EF developed in 78 (21%) patients. Of 366 patients, 103 (28%) patients died. Baseline characteristics were similar in the three groups, except female sex an indicator of abnormal EF. Mortality rates were also similar in the three groups; however, mortality rates were significantly higher in patients with abnormal EF (decreased or increased vs. normal). Echocardiographic parameters were significantly different in the three groups, in terms of LV systolic parameters and chamber size. Small left atrium (LA) and small LV were significantly associated with abnormal EF (especially in patients with increased EF). High brain natriuretic peptide was associated with decreased EF. Among these factors, female sex and small LA were significantly associated with abnormal EF in the multiple regression analysis. CONCLUSION Our findings highlight that female sex and small cardiac size are associated with abnormal EF, and therefore, death. Therefore, female patients and patients with small LA should be monitored closely when they present with sepsis.
Collapse
Affiliation(s)
- Dong Geum Shin
- Division of Cardiology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| | - Min-Kyung Kang
- Division of Cardiology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
- * E-mail:
| | - Yu Bin Seo
- Division of Infection, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| | - Jaehuk Choi
- Division of Cardiology, Dongtan Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| | - Seon Yong Choi
- Division of Cardiology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| | - Seonghoon Choi
- Division of Cardiology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| | - Jung Rae Cho
- Division of Cardiology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| | - Namho Lee
- Division of Cardiology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul, South Korea
| |
Collapse
|
42
|
Zhai T, Zhang J, Zhang Y, Wu Y. Cathelicidin deficiency exacerbates cardiac dysfunction in lipopolysaccharide-induced endotoxaemic mice. Clin Exp Pharmacol Physiol 2020; 47:677-686. [PMID: 31868940 DOI: 10.1111/1440-1681.13234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 11/30/2022]
Abstract
The therapeutic potential of the antimicrobial peptide cathelicidin (Camp) administration in sepsis has been widely investigated. However, little is known about the pathophysiological roles of cathelicidin in septic cardiomyopathy. In a lipopolysaccharide (LPS)-induced endotoxaemic model, we found that the mRNA and protein expression of cardiac cathelicidin were induced in C57BL/6J wild-type (WT) mice upon LPS challenge, accompanied by increased circulating cathelicidin levels. We showed that this peptide was mainly derived from neutrophils and monocytes/macrophages. Camp deficiency exacerbated LPS-induced myocardial depression, while the administration of CRAMP (the mature form of mouse cathelicidin) decreased the LPS-induced mortality in a D-galactosamine hydrochloride (D-GalN)-sensitized endotoxin shock model. In vivo, LPS-treated Camp knockout mice had a significant higher protein level of myocardial and circulating tumour necrosis factor-alpha (TNF-α), a major contributing factor to septic cardiomyopathy, compared to LPS-treated WT mice, while CRAMP administration inhibited LPS-induced TNF-α production in the heart and plasma in D-GalN-sensitized endotoxaemic mice. In vitro, CRAMP treatment suppressed LPS-induced Tnf-α mRNA expression in cultured neonatal mouse cardiomyocytes and reduced TNF-α secretion in the culture supernatant. The inhibitory effects of CRAMP on TNF-α production may be related to its neutralizing ability of LPS, since CRAMP application had no effects on another toll-like receptor 4 ligand paclitaxel-induced Tnf-α mRNA expression in cardiomyocytes. These findings suggest that LPS-induced cathelicidin protects the heart against myocardial depression partly through the inhibition of TNF-α production via neutralizing LPS.
Collapse
Affiliation(s)
- Tingting Zhai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Zhang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yacheng Zhang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Wu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
43
|
Duan M, Yuan Y, Liu C, Cai Z, Xie Q, Hu T, Tang Q, Wu Q. Indigo Fruits Ingredient, Aucubin, Protects against LPS-Induced Cardiac Dysfunction in Mice. J Pharmacol Exp Ther 2019; 371:348-359. [PMID: 31467086 DOI: 10.1124/jpet.119.259069] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Aucubin (AUB), which is extracted from Eucommia ulmoides Oliver seeds, has been found to possess anti-inflammatory and antiapoptotic properties. Recent studies have indicated that inflammation, oxidative stress, and apoptosis are involved in the pathophysiology of lipopolysaccharide (LPS)-induced cardiac dysfunction. Our study aimed to investigate the effect of AUB on LPS-induced acute cardiac injury. Male C57BL/6 mice were injected with LPS (one 6 mg/kg injection) to induce cardiac dysfunction without or with AUB pretreatment (20 or 80 mg/kg per day) for 1 week. We found that AUB ameliorated cardiac dysfunction, inflammation, oxidative stress, and apoptosis induced by LPS stimulation. Mechanistically, AUB inhibited LPS-induced oxidative stress by decreasing reactive oxygen species and thioredoxin interaction protein (TXNIP) levels. Moreover, AUB suppressed LPS-induced inflammation and apoptosis by reducing nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3)/apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)/caspase-1 inflammasome formation. Overexpression of NLRP3 in cardiomyocytes attenuated the protective effects of AUB. Interestingly, NLRP3 deficiency ameliorated cardiac function and reduced the inflammatory response and oxidative stress after LPS insult in mice, whereas AUB could not further prevent LPS-induced cardiac dysfunction in NLRP3-deficient mice. In summary, AUB exerts a protective effect against LPS-induced inflammation, oxidative stress, and apoptosis in vivo and in vitro by regulating the TXNIP pathway and inactivating the NLRP3/ASC/caspase-1 inflammasome. Hence, AUB may be a promising agent against LPS-induced cardiac dysfunction. SIGNIFICANCE STATEMENT: Aucubin exerts a protective effect against lipopolysaccharide-induced cardiac dysfunction by regulating nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 inflammasome.
Collapse
Affiliation(s)
- MingXia Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - Chen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - Zhulan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - Qingwen Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - Tongtong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| | - QingQing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.); and Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China (M.-X.D., Y.Y., C.L., Z.C., Q.X., T.H., Q.T., Q.-Q.W.)
| |
Collapse
|
44
|
The Role of ALDH2 in Sepsis and the To-Be-Discovered Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1193:175-194. [PMID: 31368104 DOI: 10.1007/978-981-13-6260-6_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis, defined as life-threatening tissue damage and organ dysfunction caused by a dysregulated host response to infection, is a critical disease which imposes global health burden. Sepsis-induced organ dysfunction, including circulatory and cardiac dysfunction, hepatic dysfunction, renal dysfunction, etc., contributes to high mortality and long-term disability of sepsis patients. Altered inflammatory response, ROS and reactive aldehyde stress, mitochondrial dysfunction, and programmed cell death pathways (necrosis, apoptosis, and autophagy) have been demonstrated to play crucial roles in septic organ dysfunction. Unfortunately, except for infection control and supportive therapies, no specific therapy exists for sepsis. New specific therapeutic targets are highly warranted. Emerging studies suggested a role of potential therapeutic target of ALDH2, a tetrameric enzyme located in mitochondria to detoxify aldehydes, in septic organ dysfunction. In this article, we will review the presentations and pathophysiology of septic organ dysfunction, as well as summarize and discuss the recent insights regarding ALDH2 in sepsis.
Collapse
|
45
|
Antioxidant and Cardioprotective Effects of EPA on Early Low-Severity Sepsis through UCP3 and SIRT3 Upholding of the Mitochondrial Redox Potential. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9710352. [PMID: 31534623 PMCID: PMC6732625 DOI: 10.1155/2019/9710352] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/17/2019] [Indexed: 12/29/2022]
Abstract
Sepsis still causes death, often through cardiac failure and mitochondrial dysfunction. Dietary ω3 polyunsaturated fatty acids are known to protect against cardiac dysfunction and sepsis lethality. This study set out to determine whether early low-severity sepsis alters the cardiac mitochondrial function in animals fed a Western-type diet and whether dietary eicosapentaenoic acid (EPA) administration protects the myocardium against the deleterious effects of sepsis and if so to seek possible mechanisms for its effects. Rats were divided into two groups fed either an ω3 PUFA-deficient diet (“Western diet,” DEF group) or an EPA-enriched diet (EPA group) for 5 weeks. Each group was subdivided into two subgroups: sham-operated rats and rats subjected to cecal ligation and puncture (CLP). In vivo cardiac mechanical function was examined, and mitochondria were harvested to determine their functional activity. Oxidative stress was evaluated together with several factors involved in the regulation of reactive oxygen species metabolism. Sepsis had little effect on cardiac mechanical function but strongly depressed mitochondrial function in the DEF group. Conversely, dietary EPA greatly protected the mitochondria through a decreased oxidative stress of the mitochondrial matrix. The latter was probably due to an increased uncoupling protein-3 expression, already seen in the sham-operated animals. CLP rats in the EPA group also displayed increased mitochondrial sirtuin-3 protein expression that could reinforce the upholding of oxidative phosphorylation. Dietary EPA preconditioned the heart against septic damage through several modifications that protect mitochondrial integrity. This preconditioning can explain the cardioprotective effect of dietary EPA during sepsis.
Collapse
|
46
|
Tan Y, Ouyang H, Xiao X, Zhong J, Dong M. Irisin ameliorates septic cardiomyopathy via inhibiting DRP1-related mitochondrial fission and normalizing the JNK-LATS2 signaling pathway. Cell Stress Chaperones 2019; 24:595-608. [PMID: 30993599 PMCID: PMC6527615 DOI: 10.1007/s12192-019-00992-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Irisin plays a protective effect in acute and chronic myocardial damage, but its role in septic cardiomyopathy is unclear. The aim of our study was to explore the in vivo and in vitro effects of irisin using an LPS-induced septic cardiomyopathy model. Our results demonstrated that irisin treatment attenuated LPS-mediated cardiomyocyte death and myocardial dysfunction. At the molecular level, LPS application was associated with mitochondrial oxidative injury, cardiomyocyte ATP depletion and caspase-related apoptosis activation. In contrast, the irisin treatment sustained mitochondrial function by inhibiting DRP1-related mitochondrial fission and the reactivation of mitochondrial fission impaired the protective action of irisin on inflammation-attacked mitochondria and cardiomyocytes. Additionally, we found that irisin modulated DRP1-related mitochondrial fission through the JNK-LATS2 signaling pathway. JNK activation and/or LATS2 overexpression abolished the beneficial effects of irisin on LPS-mediated mitochondrial stress and cardiomyocyte death. Altogether, our results illustrate that LPS-mediated activation of DRP1-related mitochondrial fission through the JNK-LATS2 pathway participates in the pathogenesis of septic cardiomyopathy. Irisin could be used in the future as an effective therapy for sepsis-induced myocardial depression because it corrects DRP1-related mitochondrial fission and normalizes the JNK-LATS2 signaling pathway.
Collapse
Affiliation(s)
- Ying Tan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Haichun Ouyang
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, 528300 Guangdong China
| | - Xiaochan Xiao
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Jiankai Zhong
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, 528300 Guangdong China
| | - Maolong Dong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
47
|
Tan J, Sun T, Shen J, Zhu H, Gong Y, Zhu H, Wu G. FAM46C inhibits lipopolysaccharides-induced myocardial dysfunction via downregulating cellular adhesion molecules and inhibiting apoptosis. Life Sci 2019; 229:1-12. [PMID: 30910647 DOI: 10.1016/j.lfs.2019.03.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 10/27/2022]
Abstract
AIMS Sepsis is a syndrome of inflammatory response induced by infection. Cellular adhesion molecules may involve in sepsis-induced myocardial dysfunction (SIMD) which is a major predictor of morbidity and mortality of sepsis. Here we studied the role of FAM46C in AC16 cells and c57 mice with lipopolysaccharides (LPS) treatment. MAIN METHODS Real-time PCR and western blot were used to detect the expression level of relative genes and protein. Cell proliferation and apoptosis were evaluated. KEY FINDINGS Interestingly, negative correlation between Toll-like receptor 4 (TLR4) and FAM46C in sepsis was observed. The overexpression of FAM46C reduced the apoptosis induced by LPS in AC16 cells. Inhibition of apoptosis contributed by FAM46C was mediated by adhesion molecule via blocking p38 and ERK/MAPK signaling pathway. Moreover, overexpression of Fam46c and inhibition of TLR4 by TAK-242 could attenuate apoptosis induced by LPS in vivo. SIGNIFICANCE FAM46C played an important role in SIMD via inhibiting LPS-induced myocardial dysfunction by downregulating cellular adhesion molecules and inhibiting apoptosis. It was the first time to explore the role of FAM46C in SIMD in this study.
Collapse
Affiliation(s)
- Jiaying Tan
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China
| | - Tao Sun
- Department of Cardiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China
| | - Jun Shen
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China
| | - Huigeng Zhu
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China
| | - Ye Gong
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China
| | - Hechen Zhu
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China
| | - Gang Wu
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai, PR China.
| |
Collapse
|
48
|
Sodium tanshinone IIA sulfonate attenuates cardiac dysfunction and improves survival of rats with cecal ligation and puncture-induced sepsis. Chin J Nat Med 2019; 16:846-855. [PMID: 30502766 DOI: 10.1016/s1875-5364(18)30126-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Indexed: 12/13/2022]
Abstract
Cardiac dysfunction, a common consequence of sepsis, is the major contribution to morbidity and mortality in patients. Sodium tanshinone IIA sulfonate (STS) is a water-soluble derivative of Tanshinone IIA (TA), a main active component of Salvia miltiorrhiza Bunge, which has been widely used in China for the treatment of cardiovascular and cerebral system diseases. In the present study, the effect of STS on sepsis-induced cardiac dysfunction was investigated and its effect on survival rate of rats with sepsis was also evaluated. STS treatment could significantly decrease the serum levels of C-reactive protein (CRP), procalcitonin (PCT), cardiac troponin I (cTn-I), cardiac troponin T (cTn-T), and brain natriuretic peptide (BNP) in cecal ligation and puncture (CLP)-induced) septic rats and improve left ventricular function, particularly at 48 and 72 h after CLP. As the pathogenesis of septic myocardial dysfunction is attributable to dysregulated systemic inflammatory responses, several key cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-10 (IL-10) and high mobility group protein B1 (HMGB1), were detected to reveal the possible mechanism of attenuation of septic myocardial dysfunction after being treated by STS. Our study showed that STS, especially at a high dose (15 mg·kg-1), could efficiently suppress inflammatory responses in myocardium and reduce myocardial necrosis through markedly reducing production of myocardial TNF-α, IL-6 and HMGB1. STS significantly improved the 18-day survival rate of rats with sepsis from 0% to 30% (P < 0.05). Therefore, STS could suppress inflammatory responses and improve left ventricular function in rats with sepsis, suggesting that it may be developed for the treatment of sepsis.
Collapse
|
49
|
Kim JS, Kim M, Kim YJ, Ryoo SM, Sohn CH, Ahn S, Kim WY. Troponin Testing for Assessing Sepsis-Induced Myocardial Dysfunction in Patients with Septic Shock. J Clin Med 2019; 8:jcm8020239. [PMID: 30759844 PMCID: PMC6406324 DOI: 10.3390/jcm8020239] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 01/31/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
(1) Background: Myocardial dysfunction in patients with sepsis is not an uncommon phenomenon, yet reported results are conflicting and there is no objective definition. Measurement of troponin may reflect the state of the heart and may correlate with echocardiographically derived data. This study aimed to evaluate the role of admission and peak troponin-I testing for the identification of sepsis-induced myocardial dysfunction (SIMD) by transthoracic echocardiography (TTE). (2) Methods: This was a retrospective cohort study using a prospective registry of septic shock at an Emergency Department from January 2011 and April 2017. All 1,776 consecutive adult septic shock patients treated with protocol-driven resuscitation bundle therapy and tested troponin-I were enrolled. SIMD was defined as left ventricular (LV) systolic/diastolic dysfunction, right ventricular (RV) diastolic dysfunction, or global/regional wall motion abnormalities (WMA). (3) Results: Of 660 (38.4%) septic shock patients with an elevated hs-TnI (≥0.04 ng/mL) at admission, 397 patients underwent TTE and 258 cases (65%) showed SIMD (LV systolic dysfunction (n = 163, 63.2%), LV diastolic dysfunction (n = 104, 40.3%), RV dysfunction (n = 97, 37.6%), and WMA (n = 186, 72.1%)). In multivariate analysis, peak hs-TnI (odds ratio 1.03, 95% confidence interval 1.01–1.06, p = 0.008) and ST-T wave changes in the electrocardiogram (odds ratio 1.82, 95% confidence interval 1.04–2.39, p = 0.013) were associated with SIMD, in contrast to hs-TnI level at admission. The area under the curve of peak hs-TnI was 0.668. When the peak hs-TnI cutoff value was 0.634 ng/mL, the sensitivity and specificity for SIMD were 58.6% and 59.1%, respectively. 4) Conclusions: About two-thirds of patients with an elevated hs-TnI level have various cardiac dysfunctions in terms of TTE. Rather than the initial level, the peak hs-TnI and ST-T change may be considered as a risk factor of SIMD.
Collapse
Affiliation(s)
- June-Sung Kim
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Muyeol Kim
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Youn-Jung Kim
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Seung Mok Ryoo
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Chang Hwan Sohn
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Shin Ahn
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Won Young Kim
- Department of Emergency Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| |
Collapse
|
50
|
Havaldar AA. Evaluation of sepsis induced cardiac dysfunction as a predictor of mortality. Cardiovasc Ultrasound 2018; 16:31. [PMID: 30501628 PMCID: PMC6267025 DOI: 10.1186/s12947-018-0149-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/20/2018] [Indexed: 01/21/2023] Open
Abstract
Background Sepsis is characterized by life threatening organ dysfunction with dysregulated immune response. Cardiac dysfunction seen in sepsis is unique as it is reversible within 7–10 days. Initial study by Parker et al. in 1984, showed, paradoxically lower ejection fraction in survivors of septic shock. Subsequent meta-analysis did not support that survivors had lower ejection fraction. Aim of our study was to assess the sepsis induced cardiac dysfunction by 2D echocardiography and Troponin I. Methods After obtaining institutional ethical committee approval (ref 125/2016), a prospective observational study was done in an university medical college from February 2016 to April 2016. Inclusion criteria were patients diagnosed with sepsis by new sepsis definition. Pregnant patients and patients with poor echo window were excluded. Echocardiographic assessment was done within 48 h of diagnosis of sepsis by standard methods. Primary outcome was ICU mortality and secondary outcome was ICU length of stay. Statistical analysis was done using STATA™ (Version14, College station TX). Results Fifty eight patients were screened, ten were excluded due to poor echo window. Baseline characteristics were similar in survivors and non survivors, except APACHE II, SOFA age and cumulative fluid balance. Echocardiographic parameters, mitral annular plane systolic excursion (MAPSE), E/e’ and LV systolic function assessed by visual gestalt method were found to be statistically significant. Parameters found significant in bivariate analysis were used as a covariate in logistic regression. APACHE II and MAPSE were significant co-variates in logistic regression with ROC (0.95) and calibration was satisfactory (chi2(df8),1.98, p = 0.98). Conclusions Sepsis induced cardiac dysfunction assessed by echocardiography showed measurement of MAPSE when combined with APACHE II was a good predictor of mortality. Among the echocardiographic parameters MAPSE alone was a good predictor of mortality. Results of this study need further validation from larger study.
Collapse
|